1
|
Zhao LL, Liu YJ, Guo QJ, Yan N, Yang J, Han JQ, Xie XH, Luo YS. TPM4 influences the initiation and progression of gastric cancer by modulating ferroptosis via SCD1. Clin Exp Med 2025; 25:115. [PMID: 40214825 PMCID: PMC11991984 DOI: 10.1007/s10238-025-01629-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 03/11/2025] [Indexed: 04/14/2025]
Abstract
Gastric cancer (GC) is a deadly disease with poor prognosis and few treatment options. Tropomyosin 4 (TPM4) is an actin-binding protein that stabilizes the cytoskeleton of cells and has an unclear role in GC. This study aimed to elucidate the role and underlying mechanisms of TPM4 in GC pathogenesis. The expression and diagnostic and prognostic value of TPM4 in GC were analyzed using bioinformatics. A nomogram based on TPM4 expression was created and validated with an external cohort. TPM4-knockdown GC cells and xenograft models in nude mice were used to study the function of TPM4 in vitro and in vivo. Proteomic and rescue experiments confirmed the regulatory effect of TPM4 on stearoyl-CoA desaturase 1 (SCD1) in GC. Immunohistochemistry verified the expression and correlation of the TPM4 and SCD1 proteins in GC tissues. Our study identified TPM4 as an oncogene in GC, suggesting its potential diagnostic and prognostic value. The TPM4-based nomogram showed potential prognostic value for clinical use. TPM4 knockdown inhibited GC cell proliferation, induced ferroptosis, and slowed tumor growth in vivo, which is achieved by inhibiting SCD1 expression. Immunohistochemical analysis of GC tissues revealed elevated expression levels of both TPM4 and SCD1 proteins, with a positive correlation observed between their expression. TPM4 is a promising target for new diagnostic, prognostic, and therapeutic strategies for GC. Downregulation of TPM4 inhibits GC cell growth and induces ferroptosis by suppressing SCD1 expression.
Collapse
Affiliation(s)
- Ling-Lin Zhao
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, 810001, China
- Qinghai Provincial People's Hospital, Xining, 810000, China
| | - Yu-Jun Liu
- Department of Oncology, Affiliated Hospital of Qinghai University, Xining, 810001, China
| | - Qi-Jing Guo
- Department of Oncology, Air Force Medical Center, PLA, Beijing, 100142, China
| | - Nan Yan
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, 810001, China
| | - Jie Yang
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, 810001, China
| | - Jing-Qi Han
- Department of Pathology, Affiliated Hospital of Qinghai University, Xining, 810001, China
| | - Xiao-Hong Xie
- Qinghai Provincial People's Hospital, Xining, 810000, China
| | - Yu-Shuang Luo
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, 810001, China.
- Department of Oncology, Affiliated Hospital of Qinghai University, Xining, 810001, China.
| |
Collapse
|
2
|
Yoon H, Unthank J, Pallikkuth S, Chen P, Randazzo PA. Actin Binding to the BAR Domain and Arf GAP Activity of ASAP1 Coordinately Control Actin Stress Fibers and Focal Adhesions. Biol Cell 2025; 117:e70005. [PMID: 40194952 PMCID: PMC11975550 DOI: 10.1111/boc.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/08/2025] [Accepted: 03/13/2025] [Indexed: 04/09/2025]
Abstract
BACKGROUND Actin stress fibers (SFs) and focal adhesions (FAs) are dynamic structures crucial to a range of cell behaviors including cell morphology, cell migration, proliferation, survival, and differentiation. The Arf GAP ASAP1 affects both SFs and FAs. Here, we test the hypothesis that two domains with distinct biochemical activities in ASAP1, the BAR domain that binds actin and nonmuscle myosin 2 (NM2) and the Arf GAP domain, which is necessary for inducing hydrolysis of GTP bound to Arf, coordinately regulate the structures. RESULTS We found that ASAP1 associated with bundled actin, including SFs, colocalizing with α-actinin and nonmuscle myosin 2A (NM2A), and with paxillin in FAs. Reducing ASAP1 expression altered both SFs and FAs in four cell lines that we examined. The effects of reducing ASAP1 expression could be reversed by ectopic expression of ASAP1. Reduced expression of Arf5, a substrate for ASAP1, or expression of either dominant negative or GTPase deficient mutants of Arf5, affected SFs and FAs similarly to ASAP1 knockdown. Both an active GAP domain and a BAR domain contained in the same ASAP1 polypeptide were necessary to maintain FAs and SFs. CONCLUSIONS AND SIGNIFICANCE Taken together, the results support the idea that ASAP1 coordinates the maintenance of FAs and SFs through integrated function of the BAR and GAP domains. We speculate that ASAP1 regulates SFs and their interaction with FAs through direct binding to components of the actin cytoskeleton. We discuss hypotheses related to this Arf-dependent activity of ASAP1 and propose the function of ASAP1 is not control of Arf•GTP levels.
Collapse
Affiliation(s)
- Hye‐Young Yoon
- Center for Cancer Research, National Cancer InstituteBethesdaMarylandUSA
| | - Jonah Unthank
- Center for Cancer Research, National Cancer InstituteBethesdaMarylandUSA
| | - Sandeep Pallikkuth
- Center for Cancer Research, National Cancer InstituteBethesdaMarylandUSA
| | - Pei‐Wen Chen
- Department of BiologyWilliams CollegeWilliamstownMassachusettsUSA
| | - Paul A. Randazzo
- Center for Cancer Research, National Cancer InstituteBethesdaMarylandUSA
| |
Collapse
|
3
|
Sherer LA, Nagle A, Papadaki M, Edassery S, Yoo D, D’Amico L, Brambila-Diaz D, Regnier M, Kirk JA. Calcium-Activated Sarcomere Contractility Drives Cardiomyocyte Maturation and the Response to External Mechanical Cues but is Dispensable for Sarcomere Formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.18.644054. [PMID: 40166280 PMCID: PMC11957071 DOI: 10.1101/2025.03.18.644054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Background Understanding the mechanisms of cardiomyocyte development is critical for fulfilling the potential of induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). Although myocyte development is known to depend on internal and external mechanical cues, further investigation is required to understand the contributions of different signals and how they are integrated together to generate an adult cardiomyocyte. Here, we address this gap by examining the role of calcium-activated contractility in sarcomere formation and maturation and its influence on the iPSC-CM response to nanopatterns. Methods We generated iPSCs with homozygous D65A cardiac troponin C (cTnC) mutations. This mutation prevents calcium binding to site II of cTnC, resulting in tropomyosin blocking strong myosin binding to the thin filament and inhibiting sarcomere contraction. The iPSCs were differentiated into cardiomyocytes and matured in culture over 60 days. Cells were characterized via fluorescence imaging and calcium transient analysis. WT and mutant proteomes were examined via mass spectrometry throughout differentiation and maturation. We also replated partially matured cardiomyocytes onto nanopatterned surfaces to investigate how external mechanical signals affect maturation in contractile versus non-contractile cells. Results Surprisingly, we found that sarcomeres formed in the cTnC D65A cardiomyocytes, though these sarcomeres were underdeveloped and disorganized. Mutant cardiomyocytes also exhibited significant proteomic maturation defects and abnormal calcium transients. Plating D65A cardiomyocytes on nanopatterns improved structural and proteomic maturation. However, plating WT cardiomyocytes on nanopatterns led to a reduction in sarcomeric and oxidative phosphorylation protein content. Conclusions Calcium-activated contractility is dispensable for sarcomerogenesis but critical for cardiomyocyte maturation. In non-contractile, mutant cardiomyocytes, nanopatterns enhance maturation, suggesting that external mechanical cues may partially compensate for defective contractility. However, nanopatterns did not facilitate WT maturation and may have hindered it. In addition to these novel findings, these large mass spectrometry datasets cataloging iPSC-CM maturation represent a useful resource for the cardiovascular community.
Collapse
Affiliation(s)
- Laura A. Sherer
- Department of Cell and Molecular Physiology, Loyola University Stritch School of Medicine, Maywood, IL
| | - Abigail Nagle
- Department of Bioengineering, University of Washington, Seattle, USA
| | - Mary Papadaki
- Department of Cell and Molecular Physiology, Loyola University Stritch School of Medicine, Maywood, IL
| | - Seby Edassery
- Department of Cell and Molecular Physiology, Loyola University Stritch School of Medicine, Maywood, IL
| | - Dasom Yoo
- Department of Bioengineering, University of Washington, Seattle, USA
| | - Lauren D’Amico
- Department of Bioengineering, University of Washington, Seattle, USA
| | | | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, USA
| | - Jonathan A. Kirk
- Department of Cell and Molecular Physiology, Loyola University Stritch School of Medicine, Maywood, IL
| |
Collapse
|
4
|
Kimmich MJ, Geary MA, Mi-Mi L, Votra SD, Pellenz CD, Sundaramurthy S, Pruyne D. The Sole Essential Low Molecular Weight Tropomyosin Isoform of Caenorhabditis elegans Is Essential for Pharyngeal Muscle Function. Cytoskeleton (Hoboken) 2025. [PMID: 40078096 DOI: 10.1002/cm.22014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/07/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025]
Abstract
Tropomyosin is an actin-binding protein that plays roles ranging from regulating muscle contraction to controlling cytokinesis and cell migration. The simple nematode Caenorhabditis elegans provides a useful model for studying the core functions of tropomyosin in an animal, having a relatively simple anatomy and a single tropomyosin gene, lev-11, that produces seven isoforms. Three higher molecular weight isoforms regulate the contraction of body wall and other muscles, but comparatively less is known of the functions of four lower molecular weight isoforms (LEV-11C, E, T, U). We demonstrate here that C. elegans can survive with a single low molecular weight isoform, LEV-11E. Mutants disrupted for LEV-11E die as young larvae, whereas mutants lacking all other short isoforms are viable, with no overt phenotype. Vertebrate low molecular weight tropomyosins are often considered "nonmuscle" isoforms, but we find LEV-11E localizes to sarcomeric thin filaments in pharyngeal muscle and co-precipitates from worm extracts with the formin FHOD-1, which is also associated with thin filaments in pharyngeal muscle. Pharyngeal sarcomere organization is grossly normal in larvae lacking LEV-11E, indicating that the tropomyosin is not required to stabilize thin filaments, but pharyngeal pumping is absent, suggesting LEV-11E regulates actomyosin activity similar to higher molecular weight sarcomeric tropomyosin isoforms.
Collapse
Affiliation(s)
- Michael J Kimmich
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Meaghan A Geary
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Lei Mi-Mi
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - SarahBeth D Votra
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Christopher D Pellenz
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Sumana Sundaramurthy
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - David Pruyne
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
5
|
Uyeda TQP, Yamazaki Y, Kijima ST, Noguchi TQP, Ngo KX. Multiple Mechanisms to Regulate Actin Functions: "Fundamental" Versus Lineage-Specific Mechanisms and Hierarchical Relationships. Biomolecules 2025; 15:279. [PMID: 40001582 PMCID: PMC11853071 DOI: 10.3390/biom15020279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Eukaryotic actin filaments play a central role in numerous cellular functions, with each function relying on the interaction of actin filaments with specific actin-binding proteins. Understanding the mechanisms that regulate these interactions is key to uncovering how actin filaments perform diverse roles at different cellular locations. Several distinct classes of actin regulatory mechanisms have been proposed and experimentally supported. However, these mechanisms vary in their nature and hierarchy. For instance, some operate under the control of others, highlighting hierarchical relationships. Additionally, while certain mechanisms are fundamental and ubiquitous across eukaryotes, others are lineage-specific. Here, we emphasize the fundamental importance and functional significance of the following actin regulatory mechanisms: the biochemical regulation of actin nucleators, the ATP hydrolysis-dependent aging of actin filaments, thermal fluctuation- and mechanical strain-dependent conformational changes of actin filaments, and cooperative conformational changes induced by actin-binding proteins.
Collapse
Affiliation(s)
- Taro Q. P. Uyeda
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo 169-8555, Shinjuku, Japan
| | - Yosuke Yamazaki
- RIKEN Center for Biosystems Dynamics Research, Yokohama 230-0045, Kanagawa, Japan;
| | - Saku T. Kijima
- Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba 305-8566, Ibaraki, Japan;
| | - Taro Q. P. Noguchi
- Department of Chemical Science and Engineering, National Institute of Technology, Miyakonojo College, Miyakonojo 885-0006, Miyazaki, Japan;
| | - Kien Xuan Ngo
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa 920-1192, Ishikawa, Japan;
| |
Collapse
|
6
|
Cagigas ML, Ariotti N, Hook J, Rae J, Parton RG, Bryce NS, Gunning PW, Hardeman EC. Single molecule visualization of tropomyosin isoform organization in the mammalian actin cytoskeleton. Cytoskeleton (Hoboken) 2025; 82:45-54. [PMID: 38872463 PMCID: PMC11748362 DOI: 10.1002/cm.21883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 04/22/2024] [Accepted: 05/29/2024] [Indexed: 06/15/2024]
Abstract
The actin cytoskeleton is composed of both branched and unbranched actin filaments. In mammals, the unbranched actin filaments are primarily copolymers of actin and tropomyosin. Biochemical and imaging studies indicate that different tropomyosin isoforms are segregated to different actin filament populations in cells and tissues, providing isoform-specific functionality to the actin filament. Intrinsic to this model is the prediction that single-molecule imaging of tropomyosin isoforms would confirm homopolymer formation along the length of single actin filaments, a knowledge gap that remains unaddressed in the cellular environment. We combined chemical labeling of genetically engineered tropomyosin isoforms with electron tomography to locate individual tropomyosin molecules in fibroblasts. We find that the organization of two non-muscle tropomyosins, Tpm3.1 with Tpm4.2, can be distinguished from each other using light and electron microscopy. Visualization of single tropomyosin molecules associated with actin filaments supports the hypothesis that tropomyosins form continuous homopolymers, instead of heteropolymers, in the presence of all physiologically native actin-binding proteins. This is true for both isoforms tested. Furthermore, the data suggest that the tropomyosin molecules on one side of an actin filament may not be in register with those on the opposite side, indicating that each tropomyosin polymer may assembly independently.
Collapse
Affiliation(s)
| | - Nicholas Ariotti
- School of Biomedical SciencesUNSW SydneySydneyAustralia
- Electron Microscope Unit, UNSWSydneyAustralia
- Institute for Molecular BioscienceThe University of QueenslandBrisbaneAustralia
| | - Jeff Hook
- School of Biomedical SciencesUNSW SydneySydneyAustralia
| | - James Rae
- Institute for Molecular BioscienceThe University of QueenslandBrisbaneAustralia
| | - Robert G. Parton
- Institute for Molecular BioscienceThe University of QueenslandBrisbaneAustralia
- Centre for Microscopy and MicroanalysisThe University of QueenslandBrisbaneAustralia
| | | | | | | |
Collapse
|
7
|
Macwan RS, Ferrero G, Pardini B, Naccarati A, Kozlowski PB, Papetti MJ. TPM4 overexpression drives colon epithelial cell tumorigenesis by suppressing differentiation and promoting proliferation. Neoplasia 2025; 59:101093. [PMID: 39608123 PMCID: PMC11636349 DOI: 10.1016/j.neo.2024.101093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/11/2024] [Accepted: 11/18/2024] [Indexed: 11/30/2024]
Abstract
OBJECTIVE The high morbidity and mortality associated with colorectal cancer (CRC) and the recent increases in early-onset CRC obviate the need for novel methods to detect and treat this disease, particularly at early stages. We hypothesize that aberrant expression of genes involved in the crypt-luminal migration of colon epithelial cells, a process necessary for their growth arrest and maturation, may disrupt differentiation and transition cells from a normal to tumorigenic state. METHODS We searched for contractility- and motility-related genes that are dysregulated in human CRC relative to normal colon. RNA expression of one such gene, tropomyosin 4 (TPM4), was measured by qRT-PCR and RNA-seq in human colorectal tissues at various stages of tumorigenesis: CRC, adenoma, and at-risk (grossly normal mucosa from a patient with Familial Adenomatous Polyposis, or FAP), relative to controls. Effects of aberrant TPM4 expression on colon epithelial cell proliferation and maturation were determined by overexpression using stable transfection in spontaneously differentiating Caco2 cells or silencing using siRNA in proliferating cells. RESULTS TPM4 is overexpressed at various stages of tumorigenesis, including CRC, adenoma, and grossly normal FAP colon tissue, as well as in proliferating versus differentiating Caco2 cells. TPM4.2 overexpression in differentiating Caco2 cells markedly inhibits certain aspects of maturation, notably sucrase isomaltase and glutathione-S-transferase alpha1 expression, and causes morphological and cell junction abnormalities. Conversely, siRNA-mediated suppression of TPM4.2 inhibits Caco2 proliferation. CONCLUSIONS TPM4 overexpression attenuates colon epithelial cell differentiation and promotes proliferation. Therefore, TPM4 expression may be a biomarker to enhance strategies for CRC diagnosis and treatment.
Collapse
Affiliation(s)
| | - Giulio Ferrero
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy; Department of Computer Science, University of Turin, Turin, Italy
| | - Barbara Pardini
- Italian Institute for Genomic Medicine (IIGM), c/o IRCCS Candiolo, Candiolo 10060, Turin, Italy; Candiolo Cancer Institute, FPO-IRCCS, Candiolo 10060, Turin, Italy
| | - Alessio Naccarati
- Italian Institute for Genomic Medicine (IIGM), c/o IRCCS Candiolo, Candiolo 10060, Turin, Italy; Candiolo Cancer Institute, FPO-IRCCS, Candiolo 10060, Turin, Italy
| | | | - Michael J Papetti
- Touro College of Pharmacy, New York, NY 10036, USA; Touro College of Osteopathic Medicine, New York, NY 10027, USA.
| |
Collapse
|
8
|
Dhar A, Bagyashree VT, Biswas S, Kumari J, Sridhara A, Jeevan SB, Shekhar S, Palani S. Functional redundancy and formin-independent localization of tropomyosin isoforms in Saccharomyces cerevisiae. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.04.587703. [PMID: 38617342 PMCID: PMC11014519 DOI: 10.1101/2024.04.04.587703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Tropomyosin is an actin binding protein which protects actin filaments from cofilin-mediated disassembly. Distinct tropomyosin isoforms have long been hypothesized to differentially sort to subcellular actin networks and impart distinct functionalities. Nevertheless, a mechanistic understanding of the interplay between Tpm isoforms and their functional contributions to actin dynamics has been lacking. In this study, we present and charcaterize mNeonGreen-Tpm fusion proteins that exhibit good functionality in cells as a sole copy, surpassing limitations of existing probes and enabling real-time dynamic tracking of Tpm-actin filaments in vivo. Using these functional Tpm fusion proteins, we find that S. cerevisiae Tpm isoforms, Tpm1 and Tpm2, colocalize on actin cables and indiscriminately bind to actin filaments nucleated by either formin isoform-Bnr1 and Bni1 in vivo, in contrast to the long-held paradigm of Tpm-formin pairing. We show that cellular Tpm levels regulate endocytosis by affecting balance between linear and branched actin networks in yeast cells. Finally, we discover that Tpm2 can protect and organize functional actin cables in absence of Tpm1. Overall, our work supports a concentration-dependent and formin isoform independent model of Tpm isoform binding to F-actin and demonstrates for the first time, the functional redundancy of the paralog Tpm2 in actin cable maintenance in S. cerevisiae.
Collapse
Affiliation(s)
- Anubhav Dhar
- Department of Biochemistry, Indian Institute of Science, Bengaluru, Karnataka 560012, India
- equal contribution
| | - VT Bagyashree
- Department of Biochemistry, Indian Institute of Science, Bengaluru, Karnataka 560012, India
- equal contribution
| | - Sudipta Biswas
- Departments of Physics, Cell Biology and Biochemistry, Emory University, Atlanta, GA, 30322, USA
| | - Jayanti Kumari
- Department of Biochemistry, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Amruta Sridhara
- Department of Biochemistry, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Subodh B Jeevan
- Department of Biochemistry, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Shashank Shekhar
- Departments of Physics, Cell Biology and Biochemistry, Emory University, Atlanta, GA, 30322, USA
| | - Saravanan Palani
- Department of Biochemistry, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| |
Collapse
|
9
|
Reinke PYA, Heiringhoff RS, Reindl T, Baker K, Taft MH, Meents A, Mulvihill DP, Davies OR, Fedorov R, Zahn M, Manstein DJ. Crystal structures of cables formed by the acetylated and unacetylated forms of the Schizosaccharomyces pombe tropomyosin ortholog Tpm Cdc8. J Biol Chem 2024; 300:107925. [PMID: 39461476 PMCID: PMC11626781 DOI: 10.1016/j.jbc.2024.107925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/09/2024] [Accepted: 10/17/2024] [Indexed: 10/29/2024] Open
Abstract
Cables formed by head-to-tail polymerization of tropomyosin, localized along the length of sarcomeric and cytoskeletal actin filaments, play a key role in regulating a wide range of motile and contractile processes. The stability of tropomyosin cables, their interaction with actin filaments and the functional properties of the resulting co-filaments are thought to be affected by N-terminal acetylation of tropomyosin. Here, we present high-resolution structures of cables formed by acetylated and unacetylated Schizosaccharomyces pombe tropomyosin ortholog TpmCdc8. The crystal structures represent different types of cables, each consisting of TpmCdc8 homodimers in a different conformation. The structures show how the interactions of the residues in the overlap junction contribute to cable formation and how local structural perturbations affect the conformational dynamics of the protein and its ability to transmit allosteric signals. In particular, N-terminal acetylation increases the helicity of the adjacent region, which leads to a local reduction in conformational dynamics and consequently to less fraying of the N-terminal region. This creates a more consistent complementary surface facilitating the formation of specific interactions across the overlap junction.
Collapse
Affiliation(s)
- Patrick Y A Reinke
- Institute for Biophysical Chemistry, Fritz-Hartmann-Centre for Medical Research, Hannover Medical School, Hannover, Germany; Division for Structural Biochemistry, Hannover Medical School, Hannover, Germany; FS-BMX, Deutsches Elektronen-Synchrotron DESY, Hamburg, Germany
| | - Robin S Heiringhoff
- Institute for Biophysical Chemistry, Fritz-Hartmann-Centre for Medical Research, Hannover Medical School, Hannover, Germany; Division for Structural Biochemistry, Hannover Medical School, Hannover, Germany
| | - Theresia Reindl
- Institute for Biophysical Chemistry, Fritz-Hartmann-Centre for Medical Research, Hannover Medical School, Hannover, Germany
| | - Karen Baker
- School of Biosciences, University of Kent, Canterbury, Kent, UK
| | - Manuel H Taft
- Institute for Biophysical Chemistry, Fritz-Hartmann-Centre for Medical Research, Hannover Medical School, Hannover, Germany
| | - Alke Meents
- FS-BMX, Deutsches Elektronen-Synchrotron DESY, Hamburg, Germany
| | | | - Owen R Davies
- Wellcome Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Michael Swann Building, Max Born Crescent, Edinburgh, UK
| | - Roman Fedorov
- Institute for Biophysical Chemistry, Fritz-Hartmann-Centre for Medical Research, Hannover Medical School, Hannover, Germany; Division for Structural Biochemistry, Hannover Medical School, Hannover, Germany
| | - Michael Zahn
- Institute for Biophysical Chemistry, Fritz-Hartmann-Centre for Medical Research, Hannover Medical School, Hannover, Germany; Division for Structural Biochemistry, Hannover Medical School, Hannover, Germany
| | - Dietmar J Manstein
- Institute for Biophysical Chemistry, Fritz-Hartmann-Centre for Medical Research, Hannover Medical School, Hannover, Germany; Division for Structural Biochemistry, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
10
|
Wilken MB, Fonar G, Qiu R, Bennett L, Tober J, Nations C, Pavani G, Tsao V, Garifallou J, Petit C, Maguire JA, Gagne A, Okoli N, Gadue P, Chou ST, French DL, Speck NA, Thom CS. Tropomyosin 1 deficiency facilitates cell state transitions and enhances hemogenic endothelial cell specification during hematopoiesis. Stem Cell Reports 2024; 19:1264-1276. [PMID: 39214082 PMCID: PMC11411305 DOI: 10.1016/j.stemcr.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 07/29/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Tropomyosins coat actin filaments to impact actin-related signaling and cell morphogenesis. Genome-wide association studies have linked Tropomyosin 1 (TPM1) with human blood trait variation. TPM1 has been shown to regulate blood cell formation in vitro, but it remains unclear how or when TPM1 affects hematopoiesis. Using gene-edited induced pluripotent stem cell (iPSC) model systems, we found that TPM1 knockout augmented developmental cell state transitions and key signaling pathways, including tumor necrosis factor alpha (TNF-α) signaling, to promote hemogenic endothelial (HE) cell specification and hematopoietic progenitor cell (HPC) production. Single-cell analyses revealed decreased TPM1 expression during human HE specification, suggesting that TPM1 regulated in vivo hematopoiesis via similar mechanisms. Analyses of a TPM1 gene trap mouse model showed that TPM1 deficiency enhanced HE formation during embryogenesis, without increasing the number of hematopoietic stem cells. These findings illuminate novel effects of TPM1 on developmental hematopoiesis.
Collapse
Affiliation(s)
- Madison B Wilken
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Gennadiy Fonar
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Rong Qiu
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Laura Bennett
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joanna Tober
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Catriana Nations
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Giulia Pavani
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Victor Tsao
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA; School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - James Garifallou
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Chayanne Petit
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jean Ann Maguire
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Alyssa Gagne
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nkemdilim Okoli
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA; School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul Gadue
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Stella T Chou
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Deborah L French
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nancy A Speck
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher S Thom
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
11
|
Chen A, Li S, Gui J, Zhou H, Zhu L, Mi Y. Mechanisms of tropomyosin 3 in the development of malignant tumors. Heliyon 2024; 10:e35723. [PMID: 39170461 PMCID: PMC11336884 DOI: 10.1016/j.heliyon.2024.e35723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 08/23/2024] Open
Abstract
Tropomyosin (TPM) is an important regulatory protein that binds to actin in fine myofilaments, playing a crucial role in the regulation of muscle contraction. TPM3, as one of four tropomyosin genes, is notably prevalent in eukaryotic cells. Traditionally, abnormal gene expression of TPM3 has been exclusively associated with myopathy. However, recent years have witnessed a surge in studies highlighting the close correlation between abnormal expression of TPM3 and the onset, progression, metastasis, and prognosis of various malignant tumors. In light of this, investigating the mechanisms underlying the pathogenetic role of TPM3 holds significant promise for early diagnosis and more effective treatment strategies. This article aims to provide an insightful review of the structural characteristics of TPM3 and its intricate role in the occurrence and development of malignant tumors.
Collapse
Affiliation(s)
- Anjie Chen
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu Province, China
| | - Sixin Li
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu Province, China
| | - Jiandong Gui
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu Province, China
| | - Hangsheng Zhou
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu Province, China
| | - Lijie Zhu
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
| | - Yuanyuan Mi
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
| |
Collapse
|
12
|
Vassilopoulou E, Venter C, Roth-Walter F. Malnutrition and Allergies: Tipping the Immune Balance towards Health. J Clin Med 2024; 13:4713. [PMID: 39200855 PMCID: PMC11355500 DOI: 10.3390/jcm13164713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/04/2024] [Accepted: 08/05/2024] [Indexed: 09/02/2024] Open
Abstract
Malnutrition, which includes macro- and micronutrient deficiencies, is common in individuals with allergic dermatitis, food allergies, rhinitis, and asthma. Prolonged deficiencies of proteins, minerals, and vitamins promote Th2 inflammation, setting the stage for allergic sensitization. Consequently, malnutrition, which includes micronutrient deficiencies, fosters the development of allergies, while an adequate supply of micronutrients promotes immune cells with regulatory and tolerogenic phenotypes. As protein and micronutrient deficiencies mimic an infection, the body's innate response limits access to these nutrients by reducing their dietary absorption. This review highlights our current understanding of the physiological functions of allergenic proteins, iron, and vitamin A, particularly regarding their reduced bioavailability under inflamed conditions, necessitating different dietary approaches to improve their absorption. Additionally, the role of most allergens as nutrient binders and their involvement in nutritional immunity will be briefly summarized. Their ability to bind nutrients and their close association with immune cells can trigger exaggerated immune responses and allergies in individuals with deficiencies. However, in nutrient-rich conditions, these allergens can also provide nutrients to immune cells and promote health.
Collapse
Affiliation(s)
- Emilia Vassilopoulou
- Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University, 57400 Thessaloniki, Greece
- Department of Clinical Sciences and Community Health, Univertià degli Studi die Milano, 20122 Milan, Italy
| | - Carina Venter
- Pediatrics, Section of Allergy & Immunology, University of Colorado Denver School of Medicine, Children’s Hospital Colorado, Box B518, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Franziska Roth-Walter
- Messerli Research Institute, Department of Interdisciplinary Life Sciences, University of Veterinary Medicine Vienna, Medical University of Vienna and University of Vienna, 1210 Vienna, Austria
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
13
|
Pollard LW, Boczkowska M, Dominguez R, Ostap EM. Myosin-1C differentially displaces tropomyosin isoforms altering their inhibition of motility. J Biol Chem 2024; 300:107539. [PMID: 38971309 PMCID: PMC11338116 DOI: 10.1016/j.jbc.2024.107539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/25/2024] [Accepted: 06/27/2024] [Indexed: 07/08/2024] Open
Abstract
Force generation and motility by actomyosin in nonmuscle cells are spatially regulated by ∼40 tropomyosin (Tpm) isoforms. The means by which Tpms are targeted to specific cellular regions and the mechanisms that result in differential activity of myosin paralogs are unknown. We show that Tpm3.1 and Tpm1.7 inhibit Myosin-IC (Myo1C), with Tpm1.7 more effectively reducing the number of gliding filaments than Tpm3.1. Strikingly, cosedimentation and fluorescence microscopy assays revealed that Tpm3.1 is displaced from actin by Myo1C and not by myosin-II. In contrast, Tpm1.7 is only weakly displaced by Myo1C. Unlike other characterized myosins, Myo1C motility is inhibited by Tpm when the Tpm-actin filament is activated by myosin-II. These results point to a mechanism for the exclusion of myosin-I paralogs from cellular Tpm-decorated actin filaments that are activated by other myosins. Additionally, our results suggest a potential mechanism for myosin-induced Tpm sorting in cells.
Collapse
Affiliation(s)
- Luther W Pollard
- Department of Physiology and Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| | - Malgorzata Boczkowska
- Department of Physiology and Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Roberto Dominguez
- Department of Physiology and Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - E Michael Ostap
- Department of Physiology and Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
14
|
Lapshina KK, Nefedova VV, Nabiev SR, Roman SG, Shchepkin DV, Kopylova GV, Kochurova AM, Beldiia EA, Kleymenov SY, Levitsky DI, Matyushenko AM. Functional and Structural Properties of Cytoplasmic Tropomyosin Isoforms Tpm1.8 and Tpm1.9. Int J Mol Sci 2024; 25:6873. [PMID: 38999987 PMCID: PMC11240984 DOI: 10.3390/ijms25136873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/11/2024] [Accepted: 06/19/2024] [Indexed: 07/14/2024] Open
Abstract
The actin cytoskeleton is one of the most important players in cell motility, adhesion, division, and functioning. The regulation of specific microfilament formation largely determines cellular functions. The main actin-binding protein in animal cells is tropomyosin (Tpm). The unique structural and functional diversity of microfilaments is achieved through the diversity of Tpm isoforms. In our work, we studied the properties of the cytoplasmic isoforms Tpm1.8 and Tpm1.9. The results showed that these isoforms are highly thermostable and differ in the stability of their central and C-terminal fragments. The properties of these isoforms were largely determined by the 6th exons. Thus, the strength of the end-to-end interactions, as well as the affinity of the Tpm molecule for F-actin, differed between the Tpm1.8 and Tpm1.9 isoforms. They were determined by whether an alternative internal exon, 6a or 6b, was included in the Tpm isoform structure. The strong interactions of the Tpm1.8 and Tpm1.9 isoforms with F-actin led to the formation of rigid actin filaments, the stiffness of which was measured using an optical trap. It is quite possible that the structural and functional features of the Tpm isoforms largely determine the appearance of these isoforms in the rigid actin structures of the cell cortex.
Collapse
Affiliation(s)
- Ksenia K. Lapshina
- Research Centre of Biotechnology, Russian Academy of Sciences, 119071 Moscow, Russia; (K.K.L.); (V.V.N.); (S.G.R.); (S.Y.K.); (D.I.L.)
- Department of Biophysics, Faculty of Physics, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Victoria V. Nefedova
- Research Centre of Biotechnology, Russian Academy of Sciences, 119071 Moscow, Russia; (K.K.L.); (V.V.N.); (S.G.R.); (S.Y.K.); (D.I.L.)
| | - Salavat R. Nabiev
- Institute of Immunology and Physiology, Ural Branch of Russian Academy of Sciences, 620049 Yekaterinburg, Russia; (S.R.N.); (D.V.S.); (G.V.K.); (A.M.K.); (E.A.B.)
| | - Svetlana G. Roman
- Research Centre of Biotechnology, Russian Academy of Sciences, 119071 Moscow, Russia; (K.K.L.); (V.V.N.); (S.G.R.); (S.Y.K.); (D.I.L.)
| | - Daniil V. Shchepkin
- Institute of Immunology and Physiology, Ural Branch of Russian Academy of Sciences, 620049 Yekaterinburg, Russia; (S.R.N.); (D.V.S.); (G.V.K.); (A.M.K.); (E.A.B.)
| | - Galina V. Kopylova
- Institute of Immunology and Physiology, Ural Branch of Russian Academy of Sciences, 620049 Yekaterinburg, Russia; (S.R.N.); (D.V.S.); (G.V.K.); (A.M.K.); (E.A.B.)
| | - Anastasia M. Kochurova
- Institute of Immunology and Physiology, Ural Branch of Russian Academy of Sciences, 620049 Yekaterinburg, Russia; (S.R.N.); (D.V.S.); (G.V.K.); (A.M.K.); (E.A.B.)
| | - Evgenia A. Beldiia
- Institute of Immunology and Physiology, Ural Branch of Russian Academy of Sciences, 620049 Yekaterinburg, Russia; (S.R.N.); (D.V.S.); (G.V.K.); (A.M.K.); (E.A.B.)
| | - Sergey Y. Kleymenov
- Research Centre of Biotechnology, Russian Academy of Sciences, 119071 Moscow, Russia; (K.K.L.); (V.V.N.); (S.G.R.); (S.Y.K.); (D.I.L.)
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Dmitrii I. Levitsky
- Research Centre of Biotechnology, Russian Academy of Sciences, 119071 Moscow, Russia; (K.K.L.); (V.V.N.); (S.G.R.); (S.Y.K.); (D.I.L.)
| | - Alexander M. Matyushenko
- Research Centre of Biotechnology, Russian Academy of Sciences, 119071 Moscow, Russia; (K.K.L.); (V.V.N.); (S.G.R.); (S.Y.K.); (D.I.L.)
| |
Collapse
|
15
|
Schofield MM, Rzepski AT, Richardson-Solorzano S, Hammerstedt J, Shah S, Mirack CE, Herrick M, Parreno J. Targeting F-actin stress fibers to suppress the dedifferentiated phenotype in chondrocytes. Eur J Cell Biol 2024; 103:151424. [PMID: 38823166 PMCID: PMC11610718 DOI: 10.1016/j.ejcb.2024.151424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 04/30/2024] [Accepted: 05/21/2024] [Indexed: 06/03/2024] Open
Abstract
Actin is a central mediator of the chondrocyte phenotype. Monolayer expansion of articular chondrocytes on tissue culture polystyrene, for cell-based repair therapies, leads to chondrocyte dedifferentiation. During dedifferentiation, chondrocytes spread and filamentous (F-)actin reorganizes from a cortical to a stress fiber arrangement causing a reduction in cartilage matrix expression and an increase in fibroblastic matrix and contractile molecule expression. While the downstream mechanisms regulating chondrocyte molecular expression by alterations in F-actin organization have become elucidated, the critical upstream regulators of F-actin networks in chondrocytes are not completely known. Tropomyosin (TPM) and the RhoGTPases are known regulators of F-actin networks. The main purpose of this study is to elucidate the regulation of passaged chondrocyte F-actin stress fiber networks and cell phenotype by the specific TPM, TPM3.1, and the RhoGTPase, CDC42. Our results demonstrated that TPM3.1 associates with cortical F-actin and stress fiber F-actin in primary and passaged chondrocytes, respectively. In passaged cells, we found that pharmacological TPM3.1 inhibition or siRNA knockdown causes F-actin reorganization from stress fibers back to cortical F-actin and causes an increase in G/F-actin. CDC42 inhibition also causes formation of cortical F-actin. However, pharmacological CDC42 inhibition, but not TPM3.1 inhibition, leads to the re-association of TPM3.1 with cortical F-actin. Both TPM3.1 and CDC42 inhibition, as well as TPM3.1 knockdown, reduces nuclear localization of myocardin related transcription factor, which suppresses dedifferentiated molecule expression. We confirmed that TPM3.1 or CDC42 inhibition partially redifferentiates passaged cells by reducing fibroblast matrix and contractile expression, and increasing chondrogenic SOX9 expression. A further understanding on the regulation of F-actin in passaged cells may lead into new insights to stimulate cartilage matrix expression in cells for regenerative therapies.
Collapse
Affiliation(s)
| | | | | | | | - Sohan Shah
- Department of Biological Sciences, University of Delaware, USA
| | - Chloe E Mirack
- Department of Biological Sciences, University of Delaware, USA
| | - Marin Herrick
- Department of Biological Sciences, University of Delaware, USA
| | - Justin Parreno
- Department of Biological Sciences, University of Delaware, USA; Department of Biomedical Engineering, University of Delaware, USA.
| |
Collapse
|
16
|
Wang ZX, Lin X, Cao J, Liu YW, Luo ZW, Rao SS, Wang Q, Wang YY, Chen CY, Zhu GQ, Li FXZ, Tan YJ, Hu Y, Yin H, Li YY, He ZH, Liu ZZ, Yuan LQ, Zhou Y, Wang ZG, Xie H. Young osteocyte-derived extracellular vesicles facilitate osteogenesis by transferring tropomyosin-1. J Nanobiotechnology 2024; 22:208. [PMID: 38664789 PMCID: PMC11046877 DOI: 10.1186/s12951-024-02367-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 02/22/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Bone marrow mesenchymal stem cells (BMSCs) can undergo inadequate osteogenesis or excessive adipogenesis as they age due to changes in the bone microenvironment, ultimately resulting in decreased bone density and elevated risk of fractures in senile osteoporosis. This study aims to investigate the effects of osteocyte senescence on the bone microenvironment and its influence on BMSCs during aging. RESULTS Primary osteocytes were isolated from 2-month-old and 16-month-old mice to obtain young osteocyte-derived extracellular vesicles (YO-EVs) and senescent osteocyte-derived EVs (SO-EVs), respectively. YO-EVs were found to significantly increase alkaline phosphatase activity, mineralization deposition, and the expression of osteogenesis-related genes in BMSCs, while SO-EVs promoted BMSC adipogenesis. Neither YO-EVs nor SO-EVs exerted an effect on the osteoclastogenesis of primary macrophages/monocytes. Our constructed transgenic mice, designed to trace osteocyte-derived EV distribution, revealed abundant osteocyte-derived EVs embedded in the bone matrix. Moreover, mature osteoclasts were found to release osteocyte-derived EVs from bone slices, playing a pivotal role in regulating the functions of the surrounding culture medium. Following intravenous injection into young and elderly mouse models, YO-EVs demonstrated a significant enhancement of bone mass and biomechanical strength compared to SO-EVs. Immunostaining of bone sections revealed that YO-EV treatment augmented the number of osteoblasts on the bone surface, while SO-EV treatment promoted adipocyte formation in the bone marrow. Proteomics analysis of YO-EVs and SO-EVs showed that tropomyosin-1 (TPM1) was enriched in YO-EVs, which increased the matrix stiffness of BMSCs, consequently promoting osteogenesis. Specifically, the siRNA-mediated depletion of Tpm1 eliminated pro-osteogenic activity of YO-EVs both in vitro and in vivo. CONCLUSIONS Our findings suggested that YO-EVs played a crucial role in maintaining the balance between bone resorption and formation, and their pro-osteogenic activity declining with aging. Therefore, YO-EVs and the delivered TPM1 hold potential as therapeutic targets for senile osteoporosis.
Collapse
Affiliation(s)
- Zhen-Xing Wang
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China
| | - Xiao Lin
- The Second Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jia Cao
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China
| | - Yi-Wei Liu
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, 410008, Hunan, China
| | - Zhong-Wei Luo
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, 410008, Hunan, China
| | - Shan-Shan Rao
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China
| | - Qiang Wang
- Department of Laboratory Medicine, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, 310013, Zhejiang, China
| | - Yi-Yi Wang
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, 410008, Hunan, China
| | - Chun-Yuan Chen
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China
| | - Guo-Qiang Zhu
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, 410008, Hunan, China
| | - Fu-Xing-Zi Li
- The Second Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yi-Juan Tan
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, 410008, Hunan, China
| | - Yin Hu
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Hao Yin
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, 410008, Hunan, China
| | - You-You Li
- Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Ze-Hui He
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, 410008, Hunan, China
| | - Zheng-Zhao Liu
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China
| | - Ling-Qing Yuan
- The Second Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yong Zhou
- Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Zheng-Guang Wang
- Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China.
| | - Hui Xie
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Hunan Key Laboratory of Angmedicine, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China.
| |
Collapse
|
17
|
Kumari R, Ven K, Chastney M, Kokate SB, Peränen J, Aaron J, Kogan K, Almeida-Souza L, Kremneva E, Poincloux R, Chew TL, Gunning PW, Ivaska J, Lappalainen P. Focal adhesions contain three specialized actin nanoscale layers. Nat Commun 2024; 15:2547. [PMID: 38514695 PMCID: PMC10957975 DOI: 10.1038/s41467-024-46868-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 03/13/2024] [Indexed: 03/23/2024] Open
Abstract
Focal adhesions (FAs) connect inner workings of cell to the extracellular matrix to control cell adhesion, migration and mechanosensing. Previous studies demonstrated that FAs contain three vertical layers, which connect extracellular matrix to the cytoskeleton. By using super-resolution iPALM microscopy, we identify two additional nanoscale layers within FAs, specified by actin filaments bound to tropomyosin isoforms Tpm1.6 and Tpm3.2. The Tpm1.6-actin filaments, beneath the previously identified α-actinin cross-linked actin filaments, appear critical for adhesion maturation and controlled cell motility, whereas the adjacent Tpm3.2-actin filament layer beneath seems to facilitate adhesion disassembly. Mechanistically, Tpm3.2 stabilizes ACF-7/MACF1 and KANK-family proteins at adhesions, and hence targets microtubule plus-ends to FAs to catalyse their disassembly. Tpm3.2 depletion leads to disorganized microtubule network, abnormally stable FAs, and defects in tail retraction during migration. Thus, FAs are composed of distinct actin filament layers, and each may have specific roles in coupling adhesions to the cytoskeleton, or in controlling adhesion dynamics.
Collapse
Affiliation(s)
- Reena Kumari
- HiLIFE Institute of Biotechnology, University of Helsinki, FI-00014, Helsinki, Finland
| | - Katharina Ven
- HiLIFE Institute of Biotechnology, University of Helsinki, FI-00014, Helsinki, Finland
| | - Megan Chastney
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland
| | - Shrikant B Kokate
- HiLIFE Institute of Biotechnology, University of Helsinki, FI-00014, Helsinki, Finland
| | - Johan Peränen
- HiLIFE Institute of Biotechnology, University of Helsinki, FI-00014, Helsinki, Finland
| | - Jesse Aaron
- Advanced Imaging Center, HHMI Janelia Research Campus, Ashburn, VA, 20147, USA
| | - Konstantin Kogan
- HiLIFE Institute of Biotechnology, University of Helsinki, FI-00014, Helsinki, Finland
| | - Leonardo Almeida-Souza
- HiLIFE Institute of Biotechnology, University of Helsinki, FI-00014, Helsinki, Finland
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Elena Kremneva
- HiLIFE Institute of Biotechnology, University of Helsinki, FI-00014, Helsinki, Finland
| | - Renaud Poincloux
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Teng-Leong Chew
- Advanced Imaging Center, HHMI Janelia Research Campus, Ashburn, VA, 20147, USA
| | - Peter W Gunning
- School of Biomedical Sciences, UNSW Sydney, Wallace Wurth Building, Sydney, NSW 2052, Australia
| | - Johanna Ivaska
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland
- Department of Life Technologies, University of Turku, FI-20520, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Foundation for the Finnish Cancer Institute, Tukholmankatu 8, FI-00014, Helsinki, Finland
| | - Pekka Lappalainen
- HiLIFE Institute of Biotechnology, University of Helsinki, FI-00014, Helsinki, Finland.
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
18
|
Song Y, Zhang C, Omenn GS, O’Meara MJ, Welch JD. Predicting the Structural Impact of Human Alternative Splicing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.21.572928. [PMID: 38187531 PMCID: PMC10769328 DOI: 10.1101/2023.12.21.572928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Protein structure prediction with neural networks is a powerful new method for linking protein sequence, structure, and function, but structures have generally been predicted for only a single isoform of each gene, neglecting splice variants. To investigate the structural implications of alternative splicing, we used AlphaFold2 to predict the structures of more than 11,000 human isoforms. We employed multiple metrics to identify splicing-induced structural alterations, including template matching score, secondary structure composition, surface charge distribution, radius of gyration, accessibility of post-translational modification sites, and structure-based function prediction. We identified examples of how alternative splicing induced clear changes in each of these properties. Structural similarity between isoforms largely correlated with degree of sequence identity, but we identified a subset of isoforms with low structural similarity despite high sequence similarity. Exon skipping and alternative last exons tended to increase the surface charge and radius of gyration. Splicing also buried or exposed numerous post-translational modification sites, most notably among the isoforms of BAX. Functional prediction nominated numerous functional differences among isoforms of the same gene, with loss of function compared to the reference predominating. Finally, we used single-cell RNA-seq data from the Tabula Sapiens to determine the cell types in which each structure is expressed. Our work represents an important resource for studying the structure and function of splice isoforms across the cell types of the human body.
Collapse
Affiliation(s)
- Yuxuan Song
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Chengxin Zhang
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Gilbert S. Omenn
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Matthew J. O’Meara
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Joshua D. Welch
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Department of Computer Science and Engineering, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
19
|
Schofield MM, Rzepski A, Hammerstedt J, Shah S, Mirack C, Parreno J. Targeting F-actin stress fibers to suppress the dedifferentiated phenotype in chondrocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.08.570865. [PMID: 38106134 PMCID: PMC10723437 DOI: 10.1101/2023.12.08.570865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Actin is a central mediator of the chondrocyte phenotype. Monolayer expansion of articular chondrocytes on tissue culture polystyrene, for cell-based repair therapies, leads to chondrocyte dedifferentiation. During dedifferentiation, chondrocytes spread and filamentous (F-)actin reorganizes from a cortical to a stress fiber arrangement causing a reduction in cartilage matrix expression and an increase in fibroblastic matrix and contractile molecule expression. While the downstream mechanisms regulating chondrocyte molecular expression by alterations in F-actin organization have become elucidated, the critical upstream regulators of F-actin networks in chondrocytes are not completely known. Tropomyosin (TPM) and the RhoGTPases are known regulators of F-actin networks. The purpose of this study is to elucidate the regulation of passaged chondrocyte F-actin stress fiber networks and cell phenotype by the specific TPM, TPM3.1, and the RhoGTPase, CDC42. Our results demonstrated that TPM3.1 associates with cortical F-actin and stress fiber F-actin in primary and passaged chondrocytes, respectively. In passaged cells, we found that TPM3.1 inhibition causes F-actin reorganization from stress fibers back to cortical F-actin and also causes an increase in G/F-actin. CDC42 inhibition also causes formation of cortical F-actin. However, CDC42 inhibition, but not TPM3.1 inhibition, leads to the re-association of TPM3.1 with cortical F-actin. Both TPM3.1 and CDC42 inhibition reduces nuclear localization of myocardin related transcription factor, which is known to suppress dedifferentiated molecule expression. We confirmed that TPM3.1 or CDC42 inhibition partially redifferentiates passaged cells by reducing fibroblast matrix and contractile expression, and increasing chondrogenic SOX9 expression. A further understanding on the regulation of F-actin in passaged cells may lead into new insights to stimulate cartilage matrix expression in cells for regenerative therapies.
Collapse
Affiliation(s)
| | - Alissa Rzepski
- Department of Biological Sciences, University of Delaware
| | | | - Sohan Shah
- Department of Biological Sciences, University of Delaware
| | - Chloe Mirack
- Department of Biological Sciences, University of Delaware
| | - Justin Parreno
- Department of Biological Sciences, University of Delaware
- Department of Biomedical Engineering, University of Delaware
| |
Collapse
|
20
|
Goode BL, Eskin J, Shekhar S. Mechanisms of actin disassembly and turnover. J Cell Biol 2023; 222:e202309021. [PMID: 37948068 PMCID: PMC10638096 DOI: 10.1083/jcb.202309021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/21/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023] Open
Abstract
Cellular actin networks exhibit a wide range of sizes, shapes, and architectures tailored to their biological roles. Once assembled, these filamentous networks are either maintained in a state of polarized turnover or induced to undergo net disassembly. Further, the rates at which the networks are turned over and/or dismantled can vary greatly, from seconds to minutes to hours or even days. Here, we review the molecular machinery and mechanisms employed in cells to drive the disassembly and turnover of actin networks. In particular, we highlight recent discoveries showing that specific combinations of conserved actin disassembly-promoting proteins (cofilin, GMF, twinfilin, Srv2/CAP, coronin, AIP1, capping protein, and profilin) work in concert to debranch, sever, cap, and depolymerize actin filaments, and to recharge actin monomers for new rounds of assembly.
Collapse
Affiliation(s)
- Bruce L. Goode
- Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, Waltham, MA, USA
| | - Julian Eskin
- Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, Waltham, MA, USA
| | - Shashank Shekhar
- Departments of Physics, Cell Biology and Biochemistry, Emory University, Atlanta, GA, USA
| |
Collapse
|
21
|
Alimov N, Hoeprich GJ, Padrick SB, Goode BL. Cyclase-associated protein interacts with actin filament barbed ends to promote depolymerization and formin displacement. J Biol Chem 2023; 299:105367. [PMID: 37863260 PMCID: PMC10692737 DOI: 10.1016/j.jbc.2023.105367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 10/04/2023] [Accepted: 10/12/2023] [Indexed: 10/22/2023] Open
Abstract
Cyclase-associated protein (CAP) has emerged as a central player in cellular actin turnover, but its molecular mechanisms of action are not yet fully understood. Recent studies revealed that the N terminus of CAP interacts with the pointed ends of actin filaments to accelerate depolymerization in conjunction with cofilin. Here, we use in vitro microfluidics-assisted TIRF microscopy to show that the C terminus of CAP promotes depolymerization at the opposite (barbed) ends of actin filaments. In the absence of actin monomers, full-length mouse CAP1 and C-terminal halves of CAP1 (C-CAP1) and CAP2 (C-CAP2) accelerate barbed end depolymerization. Using mutagenesis and structural modeling, we show that these activities are mediated by the WH2 and CARP domains of CAP. In addition, we observe that CAP collaborates with profilin to accelerate barbed end depolymerization and that these effects depend on their direct interaction, providing the first known example of CAP-profilin collaborative effects in regulating actin. In the presence of actin monomers, CAP1 attenuates barbed end growth and promotes formin dissociation. Overall, these findings demonstrate that CAP uses distinct domains and mechanisms to interact with opposite ends of actin filaments and drive turnover. Further, they contribute to the emerging view of actin barbed ends as sites of dynamic molecular regulation, where numerous proteins compete and cooperate with each other to tune polymer dynamics, similar to the rich complexity seen at microtubule ends.
Collapse
Affiliation(s)
- Nikita Alimov
- Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, Waltham, Massachusetts, USA
| | - Gregory J Hoeprich
- Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, Waltham, Massachusetts, USA
| | - Shae B Padrick
- Department of Biochemistry and Molecular Biology, Drexel University, Philadelphia, Pennsylvania, USA
| | - Bruce L Goode
- Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, Waltham, Massachusetts, USA.
| |
Collapse
|
22
|
Belian S, Korenkova O, Zurzolo C. Actin-based protrusions at a glance. J Cell Sci 2023; 136:jcs261156. [PMID: 37987375 DOI: 10.1242/jcs.261156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2023] Open
Abstract
Actin-based protrusions are at the base of many fundamental cellular processes, such as cell adhesion, migration and intercellular communication. In recent decades, the discovery of new types of actin-based protrusions with unique functions has enriched our comprehension of cellular processes. However, as the repertoire of protrusions continues to expand, the rationale behind the classification of newly identified and previously known structures becomes unclear. Although current nomenclature allows good categorization of protrusions based on their functions, it struggles to distinguish them when it comes to structure, composition or formation mechanisms. In this Cell Science at a Glance article, we discuss the different types of actin-based protrusions, focusing on filopodia, cytonemes and tunneling nanotubes, to help better distinguish and categorize them based on their structural and functional differences and similarities.
Collapse
Affiliation(s)
- Sevan Belian
- Institut Pasteur, Université Paris Cité, CNRS UMR 3691, Membrane Traffic and Pathogenesis, F-75015 Paris, France
- Université Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Olga Korenkova
- Institut Pasteur, Université Paris Cité, CNRS UMR 3691, Membrane Traffic and Pathogenesis, F-75015 Paris, France
- Université Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Chiara Zurzolo
- Institut Pasteur, Université Paris Cité, CNRS UMR 3691, Membrane Traffic and Pathogenesis, F-75015 Paris, France
| |
Collapse
|
23
|
Lambert MR, Gussoni E. Tropomyosin 3 (TPM3) function in skeletal muscle and in myopathy. Skelet Muscle 2023; 13:18. [PMID: 37936227 PMCID: PMC10629095 DOI: 10.1186/s13395-023-00327-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/10/2023] [Indexed: 11/09/2023] Open
Abstract
The tropomyosin genes (TPM1-4) contribute to the functional diversity of skeletal muscle fibers. Since its discovery in 1988, the TPM3 gene has been recognized as an indispensable regulator of muscle contraction in slow muscle fibers. Recent advances suggest that TPM3 isoforms hold more extensive functions during skeletal muscle development and in postnatal muscle. Additionally, mutations in the TPM3 gene have been associated with the features of congenital myopathies. The use of different in vitro and in vivo model systems has leveraged the discovery of several disease mechanisms associated with TPM3-related myopathy. Yet, the precise mechanisms by which TPM3 mutations lead to muscle dysfunction remain unclear. This review consolidates over three decades of research about the role of TPM3 in skeletal muscle. Overall, the progress made has led to a better understanding of the phenotypic spectrum in patients affected by mutations in this gene. The comprehensive body of work generated over these decades has also laid robust groundwork for capturing the multiple functions this protein plays in muscle fibers.
Collapse
Affiliation(s)
- Matthias R Lambert
- Division of Genetics and Genomics, Boston Children's Hospital, 300 Longwood Ave., Boston, MA, 02115, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA.
| | - Emanuela Gussoni
- Division of Genetics and Genomics, Boston Children's Hospital, 300 Longwood Ave., Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
- The Stem Cell Program, Boston Children's Hospital, Boston, MA, 02115, USA
| |
Collapse
|
24
|
Ono S, Watabe E, Morisaki K, Ono K, Kuroyanagi H. Alternative splicing of a single exon causes a major impact on the affinity of Caenorhabditis elegans tropomyosin isoforms for actin filaments. Front Cell Dev Biol 2023; 11:1208913. [PMID: 37745299 PMCID: PMC10512467 DOI: 10.3389/fcell.2023.1208913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 08/29/2023] [Indexed: 09/26/2023] Open
Abstract
Tropomyosin is generally known as an actin-binding protein that regulates actomyosin interaction and actin filament stability. In metazoans, multiple tropomyosin isoforms are expressed, and some of them are involved in generating subpopulations of actin cytoskeleton in an isoform-specific manner. However, functions of many tropomyosin isoforms remain unknown. Here, we report identification of a novel alternative exon in the Caenorhabditis elegans tropomyosin gene and characterization of the effects of alternative splicing on the properties of tropomyosin isoforms. Previous studies have reported six tropomyosin isoforms encoded by the C. elegans lev-11 tropomyosin gene. We identified a seventh isoform, LEV-11U, that contained a novel alternative exon, exon 7c (E7c). LEV-11U is a low-molecular-weight tropomyosin isoform that differs from LEV-11T only at the exon 7-encoded region. In silico analyses indicated that the E7c-encoded peptide sequence was unfavorable for coiled-coil formation and distinct from other tropomyosin isoforms in the pattern of electrostatic surface potentials. In vitro, LEV-11U bound poorly to actin filaments, whereas LEV-11T bound to actin filaments in a saturable manner. When these isoforms were transgenically expressed in the C. elegans striated muscle, LEV-11U was present in the diffuse cytoplasm with tendency to form aggregates, whereas LEV-11T co-localized with sarcomeric actin filaments. Worms with a mutation in E7c showed reduced motility and brood size, suggesting that this exon is important for the optimal health. These results indicate that alternative splicing of a single exon can produce biochemically diverged tropomyosin isoforms and suggest that a tropomyosin isoform with poor actin affinity has a novel biological function.
Collapse
Affiliation(s)
- Shoichiro Ono
- Departments of Pathology and Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, United States
| | - Eichi Watabe
- Laboratory of Gene Expression, Graduate School of Biomedical Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Keita Morisaki
- Departments of Pathology and Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Kanako Ono
- Departments of Pathology and Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, United States
| | - Hidehito Kuroyanagi
- Laboratory of Gene Expression, Graduate School of Biomedical Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Biochemistry, University of the Ryukyus Graduate School of Medicine, Okinawa, Japan
| |
Collapse
|
25
|
Wilken MB, Fonar G, Nations C, Pavani G, Tsao V, Garifallou J, Tober J, Bennett L, Maguire JA, Gagne A, Okoli N, Gadue P, Chou ST, Speck NA, French DL, Thom CS. Tropomyosin 1 deficiency facilitates cell state transitions to enhance hemogenic endothelial cell specification during hematopoiesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.01.555861. [PMID: 37693628 PMCID: PMC10491315 DOI: 10.1101/2023.09.01.555861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Tropomyosins coat actin filaments and impact actin-related signaling and cell morphogenesis. Genome-wide association studies have linked Tropomyosin 1 (TPM1) with human blood trait variation. Prior work suggested that TPM1 regulated blood cell formation in vitro, but it was unclear how or when TPM1 affected hematopoiesis. Using gene-edited induced pluripotent stem cell (iPSC) model systems, TPM1 knockout was found to augment developmental cell state transitions, as well as TNFα and GTPase signaling pathways, to promote hemogenic endothelial (HE) cell specification and hematopoietic progenitor cell (HPC) production. Single-cell analyses showed decreased TPM1 expression during human HE specification, suggesting that TPM1 regulated in vivo hematopoiesis via similar mechanisms. Indeed, analyses of a TPM1 gene trap mouse model showed that TPM1 deficiency enhanced the formation of HE during embryogenesis. These findings illuminate novel effects of TPM1 on developmental hematopoiesis.
Collapse
Affiliation(s)
- Madison B Wilken
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Gennadiy Fonar
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Catriana Nations
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Giulia Pavani
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Victor Tsao
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA
- School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA
| | - James Garifallou
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Joanna Tober
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Laura Bennett
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jean Ann Maguire
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Alyssa Gagne
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Nkemdilim Okoli
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA
- School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA
| | - Paul Gadue
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Stella T Chou
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Nancy A Speck
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Deborah L French
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Christopher S Thom
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
26
|
Liu B, Shen H, He J, Jin B, Tian Y, Li W, Hou L, Zhao W, Nan J, Zhao J, Shen J, Yu H, Wang Y, Shan G, Shi L, Cai X. Cytoskeleton remodeling mediated by circRNA-YBX1 phase separation suppresses the metastasis of liver cancer. Proc Natl Acad Sci U S A 2023; 120:e2220296120. [PMID: 37459535 PMCID: PMC10372620 DOI: 10.1073/pnas.2220296120] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 05/22/2023] [Indexed: 07/20/2023] Open
Abstract
Metastasis, especially intrahepatic, is a major challenge for hepatocellular carcinoma (HCC) treatment. Cytoskeleton remodeling has been identified as a vital process mediating intrahepatic spreading. Previously, we reported that HCC tumor adhesion and invasion were modulated by circular RNA (circRNA), which has emerged as an important regulator of various cellular processes and has been implicated in cancer progression. Here, we uncovered a nuclear circRNA, circASH2, which is preferentially lost in HCC tissues and inhibits HCC metastasis by altering tumor cytoskeleton structure. Tropomyosin 4 (TPM4), a critical binding protein of actin, turned out to be the major target of circASH2 and was posttranscriptionally suppressed. Such regulation is based on messenger RNA (mRNA)/precursormRNA splicing and degradation process. Furthermore, liquid-liquid phase separation of nuclear Y-box binding protein 1 (YBX1) enhanced by circASH2 augments TPM4 transcripts decay. Together, our data have revealed a tumor-suppressive circRNA and, more importantly, uncovered a fine regulation mechanism for HCC progression.
Collapse
Affiliation(s)
- Boqiang Liu
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou310016, China
- Zhejiang Provincial Key Laboratory of Laparoscopic Technology, Zhejiang University, Hangzhou310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Hangzhou310016, China
- Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou310016, China
| | - Hao Shen
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou310016, China
- Zhejiang Provincial Key Laboratory of Laparoscopic Technology, Zhejiang University, Hangzhou310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Hangzhou310016, China
- Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou310016, China
| | - Jing He
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou310016, China
- Zhejiang Provincial Key Laboratory of Laparoscopic Technology, Zhejiang University, Hangzhou310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Hangzhou310016, China
- Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou310016, China
| | - Binghan Jin
- Department of Endocrinology, The Children's Hospital, School of Medicine, National Clinical Research Center for Child Health, Zhejiang University, Hangzhou310053, China
| | - Yuanshi Tian
- Department of Diagnostic Ultrasound & Echocardiography, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou310016, China
| | - Weiqi Li
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou310016, China
- Zhejiang Provincial Key Laboratory of Laparoscopic Technology, Zhejiang University, Hangzhou310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Hangzhou310016, China
- Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou310016, China
| | - Lidan Hou
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou310016, China
- Zhejiang Provincial Key Laboratory of Laparoscopic Technology, Zhejiang University, Hangzhou310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Hangzhou310016, China
- Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou310016, China
| | - Weijun Zhao
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou310016, China
- Zhejiang Provincial Key Laboratory of Laparoscopic Technology, Zhejiang University, Hangzhou310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Hangzhou310016, China
- Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou310016, China
| | - Junjie Nan
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou310016, China
- Zhejiang Provincial Key Laboratory of Laparoscopic Technology, Zhejiang University, Hangzhou310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Hangzhou310016, China
- Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou310016, China
| | - Jia Zhao
- Zhejiang Provincial Key Laboratory of Laparoscopic Technology, Zhejiang University, Hangzhou310016, China
| | - Jiliang Shen
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou310016, China
- Zhejiang Provincial Key Laboratory of Laparoscopic Technology, Zhejiang University, Hangzhou310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Hangzhou310016, China
- Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou310016, China
| | - Hong Yu
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou310016, China
- Zhejiang Provincial Key Laboratory of Laparoscopic Technology, Zhejiang University, Hangzhou310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Hangzhou310016, China
- Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou310016, China
| | - Yifan Wang
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou310016, China
- Zhejiang Provincial Key Laboratory of Laparoscopic Technology, Zhejiang University, Hangzhou310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Hangzhou310016, China
- Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou310016, China
| | - Ge Shan
- Zhejiang University Cancer Center, Zhejiang University, Hangzhou310030, China
- Department of Pulmonary and Critical Care Medicine, Regional medical center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou310016, China
- Division of Life Science and Medicine, Department of Clinical Laboratory, First Affiliated Hospital of the University of Science and Technology of China, Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, University of Science and Technology of China, Hefei230027, China
| | - Liang Shi
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou310016, China
- Zhejiang Provincial Key Laboratory of Laparoscopic Technology, Zhejiang University, Hangzhou310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Hangzhou310016, China
- Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou310016, China
| | - Xiujun Cai
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou310016, China
- Zhejiang Provincial Key Laboratory of Laparoscopic Technology, Zhejiang University, Hangzhou310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Hangzhou310016, China
- Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou310016, China
- Zhejiang University Cancer Center, Zhejiang University, Hangzhou310030, China
| |
Collapse
|
27
|
Logvinov AS, Nefedova VV, Yampolskaya DS, Kleymenov SY, Levitsky DI, Matyushenko AM. Structural and Functional Properties of Tropomyosin Isoforms Tpm4.1 and Tpm2.1. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:801-809. [PMID: 37748876 DOI: 10.1134/s0006297923060081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/05/2023] [Accepted: 05/10/2023] [Indexed: 09/27/2023]
Abstract
Tropomyosin (Tpm) is one of the most important partners of actin filament that largely determines its properties. In animal organisms, there are different isoforms of Tpm, which are believed to be involved in the regulation of various cellular functions. However, molecular mechanisms by which various Tpm cytoplasmic regulate of the functioning of actin filaments are still poorly understood. Here, we investigated the properties of Tpm2.1 and Tpm4.1 isoforms and compared them to each other and to more extensively studied Tpm isoforms. Tpm2.1 and Tpm4.1 were very similar in their affinity to F-actin, thermal stability, and resistance to limited proteolysis by trypsin, but differed markedly in the viscosity of their solutions and thermal stability of their complexes with F-actin. The main difference of Tpm2.1 and Tpm4.1 from other Tpm isoforms (e.g., Tpm1.6 and Tpm1.7) was their extremely low thermal stability as measured by the CD and DSC methods. We suggested the possible causes of this instability based on comparing the amino acid sequences of Tpm4.1 and Tpm2.1 with the sequences of Tpm1.6 and Tpm1.7 isoforms, respectively, that have similar exon structure.
Collapse
Affiliation(s)
- Andrey S Logvinov
- Research Centre of Biotechnology, Russian Academy of Sciences, Moscow, 119071, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Victoria V Nefedova
- Research Centre of Biotechnology, Russian Academy of Sciences, Moscow, 119071, Russia
| | - Daria S Yampolskaya
- Research Centre of Biotechnology, Russian Academy of Sciences, Moscow, 119071, Russia
| | - Sergey Y Kleymenov
- Research Centre of Biotechnology, Russian Academy of Sciences, Moscow, 119071, Russia
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Dmitrii I Levitsky
- Research Centre of Biotechnology, Russian Academy of Sciences, Moscow, 119071, Russia
| | | |
Collapse
|
28
|
FGF-2 Differentially Regulates Lens Epithelial Cell Behaviour during TGF-β-Induced EMT. Cells 2023; 12:cells12060827. [PMID: 36980168 PMCID: PMC10046997 DOI: 10.3390/cells12060827] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
Fibroblast growth factor (FGF) and transforming growth factor-beta (TGF-β) can regulate and/or dysregulate lens epithelial cell (LEC) behaviour, including proliferation, fibre differentiation, and epithelial–mesenchymal transition (EMT). Earlier studies have investigated the crosstalk between FGF and TGF-β in dictating lens cell fate, that appears to be dose dependent. Here, we tested the hypothesis that a fibre-differentiating dose of FGF differentially regulates the behaviour of lens epithelial cells undergoing TGF-β-induced EMT. Postnatal 21-day-old rat lens epithelial explants were treated with a fibre-differentiating dose of FGF-2 (200 ng/mL) and/or TGF-β2 (50 pg/mL) over a 7-day culture period. We compared central LECs (CLECs) and peripheral LECs (PLECs) using immunolabelling for changes in markers for EMT (α-SMA), lens fibre differentiation (β-crystallin), epithelial cell adhesion (β-catenin), and the cytoskeleton (alpha-tropomyosin), as well as Smad2/3- and MAPK/ERK1/2-signalling. Lens epithelial explants cotreated with FGF-2 and TGF-β2 exhibited a differential response, with CLECs undergoing EMT while PLECs favoured more of a lens fibre differentiation response, compared to the TGF-β-only-treated explants where all cells in the explants underwent EMT. The CLECs cotreated with FGF and TGF-β immunolabelled for α-SMA, with minimal β-crystallin, whereas the PLECs demonstrated strong β-crystallin reactivity and little α-SMA. Interestingly, compared to the TGF-β-only-treated explants, α-SMA was significantly decreased in the CLECs cotreated with FGF/TGF-β. Smad-dependent and independent signalling was increased in the FGF-2/TGF-β2 co-treated CLECs, that had a heightened number of cells with nuclear localisation of Smad2/3 compared to the PLECs, that in contrast had more pronounced ERK1/2-signalling over Smad2/3 activation. The current study has confirmed that FGF-2 is influential in differentially regulating the behaviour of LECs during TGF-β-induced EMT, leading to a heterogenous cell population, typical of that observed in the development of post-surgical, posterior capsular opacification (PCO). This highlights the cooperative relationship between FGF and TGF-β leading to lens pathology, providing a different perspective when considering preventative measures for controlling PCO.
Collapse
|
29
|
Tang Q, Pollard LW, Homa KE, Kovar DR, Trybus KM. Acetylation of fission yeast tropomyosin does not promote differential association with cognate formins. Cytoskeleton (Hoboken) 2023; 80:77-92. [PMID: 36692369 PMCID: PMC10121778 DOI: 10.1002/cm.21745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/02/2023] [Accepted: 01/17/2023] [Indexed: 01/25/2023]
Abstract
It was proposed from cellular studies that S. pombe tropomyosin Cdc8 (Tpm) segregates into two populations due to the presence or absence of an amino-terminal acetylation that specifies which formin-mediated F-actin networks it binds, but with no supporting biochemistry. To address this mechanism in vitro, we developed methods for S. pombe actin expression in Sf9 cells. We then employed 3-color TIRF microscopy using all recombinant S. pombe proteins to probe in vitro multicomponent mechanisms involving actin, acetylated and unacetylated Tpm, formins, and myosins. Acetyl-Tpm exhibits tight binding to actin in contrast to weaker binding by unacetylated Tpm. In disagreement with the differential recruitment model, Tpm showed no preferential binding to filaments assembled by the FH1-FH2-domains of two S. pombe formins, nor did Tpm binding have any bias towards the growing formin-bound actin filament barbed end. Although our in vitro findings do not support a direct formin-tropomyosin interaction, it is possible that formins bias differential tropomyosin isoform recruitment through undiscovered mechanisms. Importantly, despite a 12% sequence divergence between skeletal and S. pombe actin, S. pombe myosins Myo2 and Myo51 exhibited similar motile behavior with these two actins, validating key prior findings with these myosins that used skeletal actin.
Collapse
Affiliation(s)
- Qing Tang
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington VT
| | - Luther W. Pollard
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington VT
| | - Kaitlin E. Homa
- Molecular Genetics and Cell Biology, Biochemistry and Molecular Biology, the University of Chicago, Chicago, IL
| | - David R. Kovar
- Molecular Genetics and Cell Biology, Biochemistry and Molecular Biology, the University of Chicago, Chicago, IL
| | - Kathleen M. Trybus
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington VT
| |
Collapse
|
30
|
Selvaraj M, Kokate SB, Reggiano G, Kogan K, Kotila T, Kremneva E, DiMaio F, Lappalainen P, Huiskonen JT. Structural basis underlying specific biochemical activities of non-muscle tropomyosin isoforms. Cell Rep 2023; 42:111900. [PMID: 36586407 DOI: 10.1016/j.celrep.2022.111900] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 11/03/2022] [Accepted: 12/07/2022] [Indexed: 12/31/2022] Open
Abstract
The actin cytoskeleton is critical for cell migration, morphogenesis, endocytosis, organelle dynamics, and cytokinesis. To support diverse cellular processes, actin filaments form a variety of structures with specific architectures and dynamic properties. Key proteins specifying actin filaments are tropomyosins. Non-muscle cells express several functionally non-redundant tropomyosin isoforms, which differentially control the interactions of other proteins, including myosins and ADF/cofilin, with actin filaments. However, the underlying molecular mechanisms have remained elusive. By determining the cryogenic electron microscopy structures of actin filaments decorated by two functionally distinct non-muscle tropomyosin isoforms, Tpm1.6 and Tpm3.2, we reveal that actin filament conformation remains unaffected upon binding. However, Tpm1.6 and Tpm3.2 follow different paths along the actin filament major groove, providing an explanation for their incapability to co-polymerize on actin filaments. We also elucidate the molecular basis underlying specific roles of Tpm1.6 and Tpm3.2 in myosin II activation and protecting actin filaments from ADF/cofilin-catalyzed severing.
Collapse
Affiliation(s)
- Muniyandi Selvaraj
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland
| | - Shrikant B Kokate
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland
| | - Gabriella Reggiano
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Konstantin Kogan
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland
| | - Tommi Kotila
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland
| | - Elena Kremneva
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland
| | - Frank DiMaio
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Pekka Lappalainen
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland.
| | - Juha T Huiskonen
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland.
| |
Collapse
|
31
|
Hanif MA, Hossen S, Lee WK, Kho KH. Molecular Characterization of Tropomyosin and Its Potential Involvement in Muscle Contraction in Pacific Abalone. Genes (Basel) 2022; 14:2. [PMID: 36672743 PMCID: PMC9858658 DOI: 10.3390/genes14010002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Tropomyosin (TPM) is a contractile protein responsible for muscle contraction through its actin-binding activity. The complete sequence of TPM in Haliotis discus hannai (Hdh-TPM) was 2160 bp, encoding 284 amino acids, and contained a TPM signature motif and a TPM domain. Gene ontology (GO) analysis based on the amino acid sequence predicted Hdh-TPM to have an actin-binding function in the cytoskeleton. The 3D analysis predicted the Hdh-TPM to have a coiled-coil α-helical structure. Phylogenetically, Hdh-TPM formed a cluster with other TPM/TPM1 proteins during analysis. The tissue-specific mRNA expression analysis found the higher expression of Hdh-TPM in the heart and muscles; however, during embryonic and larval development (ELD), the higher expression was found in the trochophore larvae and veliger larvae. Hdh-TPM expression was upregulated in fast-growing abalone. Increasing thermal stress over a long period decreased Hdh-TPM expression. Long-term starvation (>1 week) reduced the mRNA expression of Hdh-TPM in muscle; however, the mRNA expression of Hdh-TPM was significantly higher in the mantle, which may indicate overexpression. This study is the first comprehensive study to characterize the Hdh-TPM gene in Pacific abalone and to report the expression of Hdh-TPM in different organs, and during ELD, different growth patterns, thermal stress, seasonal changes, and starvation.
Collapse
Affiliation(s)
| | | | | | - Kang Hee Kho
- Department of Fisheries Science, Chonnam National University, Yeosu 59626, Republic of Korea
| |
Collapse
|
32
|
Tan B, Zeng J, Meng F, Wang S, Xiao L, Zhao X, Hong L, Zheng E, Wu Z, Li Z, Gu T. Comprehensive analysis of pre-mRNA alternative splicing regulated by m6A methylation in pig oxidative and glycolytic skeletal muscles. BMC Genomics 2022; 23:804. [PMID: 36474138 PMCID: PMC9724443 DOI: 10.1186/s12864-022-09043-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Different types of skeletal myofibers exhibit distinct physiological and metabolic properties that are associated with meat quality traits in livestock. Alternative splicing (AS) of pre-mRNA can generate multiple transcripts from an individual gene by differential selection of splice sites. N6-methyladenosine (m6A) is the most abundant modification in mRNAs, but its regulation for AS in different muscles remains unknown. RESULTS: We characterized AS events and m6A methylation pattern in pig oxidative and glycolytic muscles. A tota1 of 1294 differential AS events were identified, and differentially spliced genes were significantly enriched in processes related to different phenotypes between oxidative and glycolytic muscles. We constructed the regulatory network between splicing factors and corresponding differential AS events and identified NOVA1 and KHDRBS2 as key splicing factors. AS event was enriched in m6A-modified genes, and the methylation level was positively correlated with the number of AS events in genes. The dynamic change in m6A enrichment was associated with 115 differentially skipping exon (SE-DAS) events within 92 genes involving in various processes, including muscle contraction and myofibril assembly. We obtained 23.4% SE-DAS events (27/115) regulated by METTL3-meditaed m6A and experimentally validated the aberrant splicing of ZNF280D, PHE4DIP, and NEB. The inhibition of m6A methyltransferase METTL3 could induce the conversion of oxidative fiber to glycolytic fiber in PSCs. CONCLUSION Our study suggested that m6A modification could contribute to significant difference in phenotypes between oxidative and glycolytic muscles by mediating the regulation of AS. These findings would provide novel insights into mechanisms underlying muscle fiber conversion.
Collapse
Affiliation(s)
- Baohua Tan
- grid.20561.300000 0000 9546 5767National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China ,grid.20561.300000 0000 9546 5767Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China
| | - Jiekang Zeng
- grid.20561.300000 0000 9546 5767National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China ,grid.20561.300000 0000 9546 5767Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China
| | - Fanming Meng
- grid.135769.f0000 0001 0561 6611State Key Laboratory of Livestock and Poultry Breeding, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, 510640 Guangzhou, Guangdong People’s Republic of China
| | - Shanshan Wang
- grid.20561.300000 0000 9546 5767National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China ,grid.20561.300000 0000 9546 5767Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China
| | - Liyao Xiao
- grid.20561.300000 0000 9546 5767National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China ,grid.20561.300000 0000 9546 5767Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China
| | - Xinming Zhao
- grid.20561.300000 0000 9546 5767National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China ,grid.20561.300000 0000 9546 5767Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China
| | - Linjun Hong
- grid.20561.300000 0000 9546 5767National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China ,grid.20561.300000 0000 9546 5767Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China
| | - Enqin Zheng
- grid.20561.300000 0000 9546 5767National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China ,grid.20561.300000 0000 9546 5767Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China
| | - Zhenfang Wu
- grid.20561.300000 0000 9546 5767National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China ,grid.20561.300000 0000 9546 5767Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China ,State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, 510642 Guangzhou, China
| | - Zicong Li
- grid.20561.300000 0000 9546 5767National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China ,grid.20561.300000 0000 9546 5767Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China
| | - Ting Gu
- grid.20561.300000 0000 9546 5767National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China ,grid.20561.300000 0000 9546 5767Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China
| |
Collapse
|
33
|
Inguito KL, Schofield MM, Faghri AD, Bloom ET, Heino M, West VC, Ebron KMM, Elliott DM, Parreno J. Stress deprivation of tendon explants or Tpm3.1 inhibition in tendon cells reduces F-actin to promote a tendinosis-like phenotype. Mol Biol Cell 2022; 33:ar141. [PMID: 36129771 PMCID: PMC9727789 DOI: 10.1091/mbc.e22-02-0067] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 08/29/2022] [Accepted: 09/12/2022] [Indexed: 02/04/2023] Open
Abstract
Actin is a central mediator between mechanical force and cellular phenotype. In tendons, it is speculated that mechanical stress deprivation regulates gene expression by reducing filamentous (F)-actin. However, the mechanisms regulating tenocyte F-actin remain unclear. Tropomyosins (Tpms) are master regulators of F-actin. There are more than 40 Tpm isoforms, each having the unique capability to stabilize F-actin subpopulations. We investigated F-actin polymerization in stress-deprived tendons and tested the hypothesis that stress fiber-associated Tpm(s) stabilize F-actin to regulate cellular phenotype. Stress deprivation of mouse tail tendon down-regulated tenogenic and up-regulated protease (matrix metalloproteinase-3) mRNA levels. Concomitant with mRNA modulation were increases in G/F-actin, confirming reduced F-actin by tendon stress deprivation. To investigate the molecular regulation of F-actin, we identified that tail, Achilles, and plantaris tendons express three isoforms in common: Tpm1.6, 3.1, and 4.2. Tpm3.1 associates with F-actin in native and primary tenocytes. Tpm3.1 inhibition reduces F-actin, leading to decreases in tenogenic expression, increases in chondrogenic expression, and enhancement of protease expression in mouse and human tenocytes. These expression changes by Tpm3.1 inhibition are consistent with tendinosis progression. A further understanding of F-actin regulation in musculoskeletal cells could lead to new therapeutic interventions to prevent alterations in cellular phenotype during disease progression.
Collapse
Affiliation(s)
- Kameron L. Inguito
- Departments of Biological Sciences, University of Delaware, Newark, DE 19716
| | - Mandy M. Schofield
- Departments of Biological Sciences, University of Delaware, Newark, DE 19716
| | - Arya D. Faghri
- Departments of Biological Sciences, University of Delaware, Newark, DE 19716
| | - Ellen T. Bloom
- Biomedical Engineering, University of Delaware, Newark, DE 19716
| | - Marissa Heino
- Departments of Biological Sciences, University of Delaware, Newark, DE 19716
- Biomedical Engineering, University of Delaware, Newark, DE 19716
| | - Valerie C. West
- Biomedical Engineering, University of Delaware, Newark, DE 19716
| | | | - Dawn M. Elliott
- Biomedical Engineering, University of Delaware, Newark, DE 19716
| | - Justin Parreno
- Departments of Biological Sciences, University of Delaware, Newark, DE 19716
- Biomedical Engineering, University of Delaware, Newark, DE 19716
| |
Collapse
|
34
|
Yao M, Tijore A, Cheng D, Li JV, Hariharan A, Martinac B, Tran Van Nhieu G, Cox CD, Sheetz M. Force- and cell state-dependent recruitment of Piezo1 drives focal adhesion dynamics and calcium entry. SCIENCE ADVANCES 2022; 8:eabo1461. [PMID: 36351022 PMCID: PMC9645726 DOI: 10.1126/sciadv.abo1461] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 09/21/2022] [Indexed: 11/11/2022]
Abstract
Mechanosensing is an integral part of many physiological processes including stem cell differentiation, fibrosis, and cancer progression. Two major mechanosensing systems-focal adhesions and mechanosensitive ion channels-can convert mechanical features of the microenvironment into biochemical signals. We report here unexpectedly that the mechanosensitive calcium-permeable channel Piezo1, previously perceived to be diffusive on plasma membranes, binds to matrix adhesions in a force-dependent manner, promoting cell spreading, adhesion dynamics, and calcium entry in normal but not in most cancer cells tested except some glioblastoma lines. A linker domain in Piezo1 is needed for binding to adhesions, and overexpression of the domain blocks Piezo1 binding to adhesions, decreasing adhesion size and cell spread area. Thus, we suggest that Piezo1 is a previously unidentified component of focal adhesions in nontransformed cells that catalyzes adhesion maturation and growth through force-dependent calcium signaling, but this function is absent in most cancer cells.
Collapse
Affiliation(s)
- Mingxi Yao
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen 518055, China
- Mechanobiology Institute, National University of Singapore, Singapore 117411
- Corresponding author. (M.Y); (C.C.); (M.S.)
| | - Ajay Tijore
- Mechanobiology Institute, National University of Singapore, Singapore 117411
- Center for Biosystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India
| | - Delfine Cheng
- Victor Chang Cardiac Research Institute, Sydney NSW 2010, Australia
| | - Jinyuan Vero Li
- Victor Chang Cardiac Research Institute, Sydney NSW 2010, Australia
| | - Anushya Hariharan
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Boris Martinac
- Victor Chang Cardiac Research Institute, Sydney NSW 2010, Australia
| | - Guy Tran Van Nhieu
- Ecole Normale Supérieure Paris-Saclay Gif-sur-Yvette, France
- Team Ca Signaling and Microbial Infections, Institute for Integrative Biology of the Cell–CNRS UMR9198–Inserm U1280, 1, Avenue de la Terrasse, 91190 Gif-sur-Yvette, France
| | - Charles D. Cox
- Victor Chang Cardiac Research Institute, Sydney NSW 2010, Australia
- Corresponding author. (M.Y); (C.C.); (M.S.)
| | - Michael Sheetz
- Mechanobiology Institute, National University of Singapore, Singapore 117411
- Department of Biological Sciences, National University of Singapore, Singapore 117558
- Molecular MechanoMedicine Program, Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
- Corresponding author. (M.Y); (C.C.); (M.S.)
| |
Collapse
|
35
|
Cowan JM, Duggan JJ, Hewitt BR, Petrie RJ. Non-muscle myosin II and the plasticity of 3D cell migration. Front Cell Dev Biol 2022; 10:1047256. [PMID: 36438570 PMCID: PMC9691290 DOI: 10.3389/fcell.2022.1047256] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 10/31/2022] [Indexed: 09/08/2024] Open
Abstract
Confined cells migrating through 3D environments are also constrained by the laws of physics, meaning for every action there must be an equal and opposite reaction for cells to achieve motion. Fascinatingly, there are several distinct molecular mechanisms that cells can use to move, and this is reflected in the diverse ways non-muscle myosin II (NMII) can generate the mechanical forces necessary to sustain 3D cell migration. This review summarizes the unique modes of 3D migration, as well as how NMII activity is regulated and localized within each of these different modes. In addition, we highlight tropomyosins and septins as two protein families that likely have more secrets to reveal about how NMII activity is governed during 3D cell migration. Together, this information suggests that investigating the mechanisms controlling NMII activity will be helpful in understanding how a single cell transitions between distinct modes of 3D migration in response to the physical environment.
Collapse
Affiliation(s)
| | | | | | - Ryan J. Petrie
- Department of Biology, Drexel University, Philadelphia, PA, United States
| |
Collapse
|
36
|
Kokate SB, Ciuba K, Tran VD, Kumari R, Tojkander S, Engel U, Kogan K, Kumar S, Lappalainen P. Caldesmon controls stress fiber force-balance through dynamic cross-linking of myosin II and actin-tropomyosin filaments. Nat Commun 2022; 13:6032. [PMID: 36229430 PMCID: PMC9561149 DOI: 10.1038/s41467-022-33688-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 09/28/2022] [Indexed: 11/09/2022] Open
Abstract
Contractile actomyosin bundles are key force-producing and mechanosensing elements in muscle and non-muscle tissues. Whereas the organization of muscle myofibrils and mechanism regulating their contractility are relatively well-established, the principles by which myosin-II activity and force-balance are regulated in non-muscle cells have remained elusive. We show that Caldesmon, an important component of smooth muscle and non-muscle cell actomyosin bundles, is an elongated protein that functions as a dynamic cross-linker between myosin-II and tropomyosin-actin filaments. Depletion of Caldesmon results in aberrant lateral movement of myosin-II filaments along actin bundles, leading to irregular myosin distribution within stress fibers. This manifests as defects in stress fiber network organization and contractility, and accompanied problems in cell morphogenesis, migration, invasion, and mechanosensing. These results identify Caldesmon as critical factor that ensures regular myosin-II spacing within non-muscle cell actomyosin bundles, and reveal how stress fiber networks are controlled through dynamic cross-linking of tropomyosin-actin and myosin filaments.
Collapse
Affiliation(s)
- Shrikant B Kokate
- HiLIFE Institute of Biotechnology, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Katarzyna Ciuba
- HiLIFE Institute of Biotechnology, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland.,Nencki Institute of Experimental Biology PAS, 3 Pasteur Street, 02-093, Warszawa, Poland
| | - Vivien D Tran
- Department of Bioengineering, University of California, Berkeley, CA, 94720, USA
| | - Reena Kumari
- HiLIFE Institute of Biotechnology, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Sari Tojkander
- Faculty of Medicine and Health Technology, Tampere University, Kauppi Campus, Arvo Building, E318, Arvo Ylpön katu 34, 33520, Tampere, Finland
| | - Ulrike Engel
- Nikon Imaging Center at Heidelberg University and Centre for Organismal Studies (COS), Heidelberg University, Im Neuenheimer Feld 267, Heidelberg, 69120, Germany
| | - Konstantin Kogan
- HiLIFE Institute of Biotechnology, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Sanjay Kumar
- Department of Bioengineering, University of California, Berkeley, CA, 94720, USA
| | - Pekka Lappalainen
- HiLIFE Institute of Biotechnology, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland.
| |
Collapse
|
37
|
Chung J, Goode BL, Gelles J. Single-molecule analysis of actin filament debranching by cofilin and GMF. Proc Natl Acad Sci U S A 2022; 119:e2115129119. [PMID: 35858314 PMCID: PMC9304009 DOI: 10.1073/pnas.2115129119] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 05/31/2022] [Indexed: 01/13/2023] Open
Abstract
Eukaryotic cells contain branched actin networks that are essential for endocytosis, motility, and other key cellular processes. These networks, which are formed by filamentous actin and the Arp2/3 complex, must subsequently be debranched to allow network remodeling and to recycle the Arp2/3 complex. Debranching appears to be catalyzed by two different members of the actin depolymerizing factor homology protein family: cofilin and glial maturation factor (GMF). However, their mechanisms of debranching are only partially understood. Here, we used single-molecule fluorescence imaging of Arp2/3 complex and actin filaments under physiological ionic conditions to observe debranching by GMF and cofilin. We demonstrate that cofilin, like GMF, is an authentic debrancher independent of its filament-severing activity and that the debranching activities of the two proteins are additive. While GMF binds directly to the Arp2/3 complex, cofilin selectively accumulates on branch-junction daughter filaments in tropomyosin-decorated networks just prior to debranching events. Quantitative comparison of debranching rates with the known kinetics of cofilin-actin binding suggests that cofilin occupancy of a particular single actin site at the branch junction is sufficient to trigger debranching. In rare cases in which the order of departure could be resolved during GMF- or cofilin-induced debranching, the Arp2/3 complex left the branch junction bound to the pointed end of the daughter filament, suggesting that both GMF and cofilin can work by destabilizing the mother filament-Arp2/3 complex interface. Taken together, these observations suggest that GMF and cofilin promote debranching by distinct yet complementary mechanisms.
Collapse
Affiliation(s)
- Johnson Chung
- Department of Biochemistry, Brandeis University, Waltham, MA 02454
| | - Bruce L. Goode
- Department of Biology, Brandeis University, Waltham, MA 02454
| | - Jeff Gelles
- Department of Biochemistry, Brandeis University, Waltham, MA 02454
| |
Collapse
|
38
|
Reindl T, Giese S, Greve JN, Reinke PY, Chizhov I, Latham SL, Mulvihill DP, Taft MH, Manstein DJ. Distinct actin–tropomyosin cofilament populations drive the functional diversification of cytoskeletal myosin motor complexes. iScience 2022; 25:104484. [PMID: 35720262 PMCID: PMC9204724 DOI: 10.1016/j.isci.2022.104484] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/02/2022] [Accepted: 05/24/2022] [Indexed: 11/02/2022] Open
Abstract
The effects of N-terminal acetylation of the high molecular weight tropomyosin isoforms Tpm1.6 and Tpm2.1 and the low molecular weight isoforms Tpm1.12, Tpm3.1, and Tpm4.2 on the actin affinity and the thermal stability of actin-tropomyosin cofilaments are described. Furthermore, we show how the exchange of cytoskeletal tropomyosin isoforms and their N-terminal acetylation affects the kinetic and chemomechanical properties of cytoskeletal actin-tropomyosin-myosin complexes. Our results reveal the extent to which the different actin-tropomyosin-myosin complexes differ in their kinetic and functional properties. The maximum sliding velocity of the actin filament as well as the optimal motor density for continuous unidirectional movement, parameters that were previously considered to be unique and invariant properties of each myosin isoform, are shown to be influenced by the exchange of the tropomyosin isoform and the N-terminal acetylation of tropomyosin. Tpm diversity is largely determined by sequences contributing to the overlap region Global sequence differences are of greater importance than variable exon 6 usage Tpm isoforms confer distinctly altered properties to cytoskeletal myosin motors Cytoskeletal myosins are differentially affected by N-terminal acetylation of Tpm
Collapse
|
39
|
Vakhrusheva A, Murashko A, Trifonova E, Efremov Y, Timashev P, Sokolova O. Role of Actin-binding Proteins in the Regulation of Cellular Mechanics. Eur J Cell Biol 2022; 101:151241. [DOI: 10.1016/j.ejcb.2022.151241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 04/18/2022] [Accepted: 05/19/2022] [Indexed: 12/25/2022] Open
|
40
|
Atoum MF, Alzoughool FE, Al-Mazaydeh ZA, Rammaha MS, Tahtamouni LH. Vitamin B12 enhances the antitumor activity of 1,25-dihydroxyvitamin D3 via activation of caspases and targeting actin cytoskeleton. Tumour Biol 2022; 44:17-35. [PMID: 35180142 DOI: 10.3233/tub-211536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND 1,25-dihydroxyvitamin D3 (1,25(OH)2D3) is an effective anticancer agent, and when combined with other agents it shows superior activities. Vitamin B12 has been shown to contribute to increasing the effectiveness of anticancer drugs when used in combination. Thus, the current study aimed at investigating the anticancer potential of the combination of 1,25(OH)2D3 and vitamin B12. METHODS MTT assay was used to determine the cytotoxic activity of combining 1,25(OH)2D3 and vitamin B12 against six different cancer cell lines and one normal cell line. The surviving fraction after clonogenic assay was measured, and the effects of 1,25(OH)2D3/B12 combination on the activity of different caspases, cell adhesion, actin cytoskeleton, cell morphology, and percentage of polarized cells were evaluated. RESULTS Vitamin B12 did not cause cytotoxicity, however, it enhanced the cytotoxicity of 1,25(OH)2D3 against cancer cells. The cytotoxic effects of 1,25(OH)2D3 and its combination with vitamin B12 was not evident in the normal mammary MCF10A cell line indicating cancer cell-specificity. The cytotoxic effects of 1,25(OH)2D3/B12 combination occurred in a dose-dependent manner and was attributed to apoptosis induction which was mediated by caspase 4 and 8. Moreover, 1,25(OH)2D3/B12-treated cells showed enhanced inhibition of clonogenic tumor growth, reduced cell adhesion, reduced cell area, reduced percentage of cell polarization, and disorganized actin cytoskeleton resulting in reduced migratory phenotype when compared to cells treated with 1,25(OH)2D3 alone. CONCLUSION 1,25(OH)2D3 and vitamin B12 exhibited synergistic anticancer effects against different cancer cell lines. The combination therapy of 1,25(OH)2D3 and vitamin B12 may provide a potential adjunctive treatment option for some cancer types.
Collapse
Affiliation(s)
- Manar F Atoum
- Department Medical Laboratory Sciences, Faculty Applied Health Sciences, The Hashemite University, Zarqa, Jordan
| | - Foad E Alzoughool
- Department Medical Laboratory Sciences, Faculty Applied Health Sciences, The Hashemite University, Zarqa, Jordan.,Faculty of Health Sciences, Fujairah Women's College, Higher Colleges Technology, UAE
| | - Zainab A Al-Mazaydeh
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Majdoleen S Rammaha
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Lubna H Tahtamouni
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan.,Department of Biochemistry and Molecular Biology, College of Natural Sciences, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
41
|
Stapley RJ, Poulter NS, Khan AO, Smith CW, Bignell P, Fratter C, Lester W, Lowe G, Morgan NV. Rare missense variants in Tropomyosin-4 (TPM4) are associated with platelet dysfunction, cytoskeletal defects, and excessive bleeding. J Thromb Haemost 2022; 20:478-485. [PMID: 34758189 DOI: 10.1111/jth.15584] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/01/2021] [Accepted: 11/05/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND A significant challenge is faced for the genetic diagnosis of inherited platelet disorders in which candidate genetic variants can be found in more than 100 bleeding, thrombotic, and platelet disorder genes, especially within families in which there are both normal and low platelet counts. Genetic variants of unknown clinical significance (VUS) are found in a significant proportion of such patients in which functional studies are required to prove pathogenicity. OBJECTIVE To identify the genetic cause in patients with a suspected platelet disorder and subsequently perform a detailed functional analysis of the candidate genetic variants found. METHODS Genetic and functional studies were undertaken in three patients in two unrelated families with a suspected platelet disorder and excessive bleeding. A targeted gene panel of previously known bleeding and platelet genes was used to identify plausible genetic variants. Deep platelet phenotyping was performed using platelet spreading analysis, transmission electron microscopy, immunofluorescence, and platelet function testing using lumiaggregometry and flow cytometry. RESULTS We report rare conserved missense variants (p.R182C and p.A183V) in TPM4 encoding tromomyosin-4 in 3 patients. Deep platelet phenotyping studies revealed similar platelet function defects across the 3 patients including reduced platelet secretion, and aggregation and spreading defects suggesting that TPM4 missense variants impact platelet function and show a disordered pattern of tropomyosin staining. CONCLUSIONS Genetic and functional TPM4 defects are reported making TPM4 a diagnostic grade tier 1 gene and highlights the importance of including TPM4 in diagnostic genetic screening for patients with significant bleeding and undiagnosed platelet disorders, particularly for those with a normal platelet count.
Collapse
Affiliation(s)
- Rachel J Stapley
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Natalie S Poulter
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Midlands, UK
| | - Abdullah O Khan
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Christopher W Smith
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Patricia Bignell
- Oxford Genetics Laboratories, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Carl Fratter
- Oxford Genetics Laboratories, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Will Lester
- Comprehensive Care Haemophilia Centre, University Hospital Birmingham NHS Foundation Trust, Birmingham, UK
| | - Gillian Lowe
- Comprehensive Care Haemophilia Centre, University Hospital Birmingham NHS Foundation Trust, Birmingham, UK
| | - Neil V Morgan
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
42
|
Abstract
The precise assembly and disassembly of actin filaments is required for several cellular processes, and their regulation has been scrutinized for decades. Twenty years ago, a handful of studies marked the advent of a new type of experiment to study actin dynamics: using optical microscopy to look at individual events, taking place on individual filaments in real time. Here, we summarize the main characteristics of this approach and how it has changed our ability to understand actin assembly dynamics. We also highlight some of its caveats and reflect on what we have learned over the past 20 years, leading us to propose a set of guidelines, which we hope will contribute to a better exploitation of this powerful tool.
Collapse
|
43
|
Verdile V, Guizzo G, Ferrante G, Paronetto MP. RNA Targeting in Inherited Neuromuscular Disorders: Novel Therapeutic Strategies to Counteract Mis-Splicing. Cells 2021; 10:2850. [PMID: 34831073 PMCID: PMC8616048 DOI: 10.3390/cells10112850] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/08/2021] [Accepted: 10/18/2021] [Indexed: 01/14/2023] Open
Abstract
Neuromuscular disorders represent multifaceted abnormal conditions, with little or no cure, leading to patient deaths from complete muscle wasting and atrophy. Despite strong efforts in the past decades, development of effective treatments is still urgently needed. Advent of next-generation sequencing technologies has allowed identification of novel genes and mutations associated with neuromuscular pathologies, highlighting splicing defects as essential players. Deciphering the significance and relative contributions of defective RNA metabolism will be instrumental to address and counteract these malignancies. We review here recent progress on the role played by alternative splicing in ensuring functional neuromuscular junctions (NMJs), and its involvement in the pathogenesis of NMJ-related neuromuscular disorders, with particular emphasis on congenital myasthenic syndromes and muscular dystrophies. We will also discuss novel strategies based on oligonucleotides designed to bind their cognate sequences in the RNA or targeting intermediary of mRNA metabolism. These efforts resulted in several chemical classes of RNA molecules that have recently proven to be clinically effective, more potent and better tolerated than previous strategies.
Collapse
Affiliation(s)
- Veronica Verdile
- Laboratory of Molecular and Cellular Neurobiology, Fondazione Santa Lucia, CERC, 00143 Rome, Italy; (V.V.); (G.G.); (G.F.)
- Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 6, 00135 Rome, Italy
| | - Gloria Guizzo
- Laboratory of Molecular and Cellular Neurobiology, Fondazione Santa Lucia, CERC, 00143 Rome, Italy; (V.V.); (G.G.); (G.F.)
| | - Gabriele Ferrante
- Laboratory of Molecular and Cellular Neurobiology, Fondazione Santa Lucia, CERC, 00143 Rome, Italy; (V.V.); (G.G.); (G.F.)
| | - Maria Paola Paronetto
- Laboratory of Molecular and Cellular Neurobiology, Fondazione Santa Lucia, CERC, 00143 Rome, Italy; (V.V.); (G.G.); (G.F.)
- Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 6, 00135 Rome, Italy
| |
Collapse
|
44
|
Bamburg JR, Minamide LS, Wiggan O, Tahtamouni LH, Kuhn TB. Cofilin and Actin Dynamics: Multiple Modes of Regulation and Their Impacts in Neuronal Development and Degeneration. Cells 2021; 10:cells10102726. [PMID: 34685706 PMCID: PMC8534876 DOI: 10.3390/cells10102726] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 02/06/2023] Open
Abstract
Proteins of the actin depolymerizing factor (ADF)/cofilin family are ubiquitous among eukaryotes and are essential regulators of actin dynamics and function. Mammalian neurons express cofilin-1 as the major isoform, but ADF and cofilin-2 are also expressed. All isoforms bind preferentially and cooperatively along ADP-subunits in F-actin, affecting the filament helical rotation, and when either alone or when enhanced by other proteins, promotes filament severing and subunit turnover. Although self-regulating cofilin-mediated actin dynamics can drive motility without post-translational regulation, cells utilize many mechanisms to locally control cofilin, including cooperation/competition with other proteins. Newly identified post-translational modifications function with or are independent from the well-established phosphorylation of serine 3 and provide unexplored avenues for isoform specific regulation. Cofilin modulates actin transport and function in the nucleus as well as actin organization associated with mitochondrial fission and mitophagy. Under neuronal stress conditions, cofilin-saturated F-actin fragments can undergo oxidative cross-linking and bundle together to form cofilin-actin rods. Rods form in abundance within neurons around brain ischemic lesions and can be rapidly induced in neurites of most hippocampal and cortical neurons through energy depletion or glutamate-induced excitotoxicity. In ~20% of rodent hippocampal neurons, rods form more slowly in a receptor-mediated process triggered by factors intimately connected to disease-related dementias, e.g., amyloid-β in Alzheimer’s disease. This rod-inducing pathway requires a cellular prion protein, NADPH oxidase, and G-protein coupled receptors, e.g., CXCR4 and CCR5. Here, we will review many aspects of cofilin regulation and its contribution to synaptic loss and pathology of neurodegenerative diseases.
Collapse
Affiliation(s)
- James R. Bamburg
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA; (L.S.M.); (O.W.); (L.H.T.); (T.B.K.)
- Correspondence: ; Tel.: +1-970-988-9120; Fax: +1-970-491-0494
| | - Laurie S. Minamide
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA; (L.S.M.); (O.W.); (L.H.T.); (T.B.K.)
| | - O’Neil Wiggan
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA; (L.S.M.); (O.W.); (L.H.T.); (T.B.K.)
| | - Lubna H. Tahtamouni
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA; (L.S.M.); (O.W.); (L.H.T.); (T.B.K.)
- Department of Biology and Biotechnology, The Hashemite University, Zarqa 13115, Jordan
| | - Thomas B. Kuhn
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA; (L.S.M.); (O.W.); (L.H.T.); (T.B.K.)
- Department of Chemistry and Biochemistry, University of Alaska, Fairbanks, AK 99775, USA
| |
Collapse
|
45
|
Carman PJ, Barrie KR, Dominguez R. Novel human cell expression method reveals the role and prevalence of posttranslational modification in nonmuscle tropomyosins. J Biol Chem 2021; 297:101154. [PMID: 34478714 PMCID: PMC8463859 DOI: 10.1016/j.jbc.2021.101154] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 08/25/2021] [Accepted: 08/30/2021] [Indexed: 11/29/2022] Open
Abstract
Biochemical studies require large quantities of proteins, which are typically obtained using bacterial overexpression. However, the folding machinery in bacteria is inadequate for expressing many mammalian proteins, which additionally undergo posttranslational modifications (PTMs) that bacteria, yeast, or insect cells cannot perform. Many proteins also require native N- and C-termini and cannot tolerate extra tag amino acids for proper function. Tropomyosin (Tpm), a coiled coil protein that decorates most actin filaments in cells, requires both native N- and C-termini and PTMs, specifically N-terminal acetylation (Nt-acetylation), to polymerize along actin filaments. Here, we describe a new method that combines native protein expression in human cells with an intein-based purification tag that can be precisely removed after purification. Using this method, we expressed several nonmuscle Tpm isoforms (Tpm1.6, Tpm1.7, Tpm2.1, Tpm3.1, Tpm3.2, and Tpm4.2) and the muscle isoform Tpm1.1. Proteomics analysis revealed that human-cell-expressed Tpms present various PTMs, including Nt-acetylation, Ser/Thr phosphorylation, Tyr phosphorylation, and Lys acetylation. Depending on the Tpm isoform (humans express up to 40 Tpm isoforms), Nt-acetylation occurs on either the initiator methionine or on the second residue after removal of the initiator methionine. Human-cell-expressed Tpms bind F-actin differently than their Escherichia coli-expressed counterparts, with or without N-terminal extensions intended to mimic Nt-acetylation, and they can form heterodimers in cells and in vitro. The expression method described here reveals previously unknown features of nonmuscle Tpms and can be used in future structural and biochemical studies with Tpms and other proteins, as shown here for α-synuclein.
Collapse
Affiliation(s)
- Peter J Carman
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kyle R Barrie
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Roberto Dominguez
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
46
|
Caporali S, Calabrese C, Minieri M, Pieri M, Tarantino U, Marini M, D’Ottavio S, Angeletti S, Mauriello A, Cortese C, Bernardini S, Terrinoni A. The miR-133a, TPM4 and TAp63γ Role in Myocyte Differentiation Microfilament Remodelling and Colon Cancer Progression. Int J Mol Sci 2021; 22:ijms22189818. [PMID: 34575979 PMCID: PMC8472330 DOI: 10.3390/ijms22189818] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/01/2021] [Accepted: 09/05/2021] [Indexed: 01/16/2023] Open
Abstract
MicroRNAs (miRNAs) play an essential role in the regulation of a number of physiological functions. miR-133a and other muscular miRs (myomiRs) play a key role in muscle cell growth and in some type of cancers. Here, we show that miR133a is upregulated in individuals that undertake physical exercise. We used a skeletal muscle differentiation model to dissect miR-133a's role and to identify new targets, identifying Tropomyosin-4 (TPM4). This protein is expressed during muscle differentiation, but importantly it is an essential component of microfilament cytoskeleton and stress fibres formation. The microfilament scaffold remodelling is an essential step in cell transformation and tumour progression. Using the muscle system, we obtained valuable information about the microfilament proteins, and the knowledge on these molecular players can be transferred to the cytoskeleton rearrangement observed in cancer cells. Further investigations showed a role of TPM4 in cancer physiology, specifically, we found that miR-133a downregulation leads to TPM4 upregulation in colon carcinoma (CRC), and this correlates with a lower patient survival. At molecular level, we demonstrated in myocyte differentiation that TPM4 is positively regulated by the TA isoform of the p63 transcription factor. In muscles, miR-133a generates a myogenic stimulus, reducing the differentiation by downregulating TPM4. In this system, miR-133a counteracts the differentiative TAp63 activity. Interestingly, in CRC cell lines and in patient biopsies, miR-133a is able to regulate TPM4 activity, while TAp63 is not active. The downregulation of the miR leads to TPM4 overexpression, this modifies the architecture of the cell cytoskeleton contributing to increase the invasiveness of the tumour and associating with a poor prognosis. These results add data to the interesting question about the link between physical activity, muscle physiology and protection against colorectal cancer. The two phenomena have in common the cytoskeleton remodelling, due to the TPM4 activity, that is involved in stress fibres formation.
Collapse
Affiliation(s)
- Sabrina Caporali
- Department of Industrial Engineering, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Cosimo Calabrese
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (C.C.); (M.M.); (M.P.); (A.M.); (C.C.); (S.B.)
| | - Marilena Minieri
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (C.C.); (M.M.); (M.P.); (A.M.); (C.C.); (S.B.)
| | - Massimo Pieri
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (C.C.); (M.M.); (M.P.); (A.M.); (C.C.); (S.B.)
| | - Umberto Tarantino
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (U.T.); (S.D.)
| | - Mario Marini
- Centre of Space Biomedicine and Department of Systems Medicine of the University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Stefano D’Ottavio
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (U.T.); (S.D.)
| | - Silvia Angeletti
- Unit of Clinical Laboratory Science, University Campus Bio-Medico of Rome, Via Alvaro del Portillo, 00128 Rome, Italy;
| | - Alessandro Mauriello
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (C.C.); (M.M.); (M.P.); (A.M.); (C.C.); (S.B.)
| | - Claudio Cortese
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (C.C.); (M.M.); (M.P.); (A.M.); (C.C.); (S.B.)
| | - Sergio Bernardini
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (C.C.); (M.M.); (M.P.); (A.M.); (C.C.); (S.B.)
| | - Alessandro Terrinoni
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (C.C.); (M.M.); (M.P.); (A.M.); (C.C.); (S.B.)
- Correspondence:
| |
Collapse
|
47
|
Kadzik RS, Homa KE, Kovar DR. F-Actin Cytoskeleton Network Self-Organization Through Competition and Cooperation. Annu Rev Cell Dev Biol 2021; 36:35-60. [PMID: 33021819 DOI: 10.1146/annurev-cellbio-032320-094706] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Many fundamental cellular processes such as division, polarization, endocytosis, and motility require the assembly, maintenance, and disassembly of filamentous actin (F-actin) networks at specific locations and times within the cell. The particular function of each network is governed by F-actin organization, size, and density as well as by its dynamics. The distinct characteristics of different F-actin networks are determined through the coordinated actions of specific sets of actin-binding proteins (ABPs). Furthermore, a cell typically assembles and uses multiple F-actin networks simultaneously within a common cytoplasm, so these networks must self-organize from a common pool of shared globular actin (G-actin) monomers and overlapping sets of ABPs. Recent advances in multicolor imaging and analysis of ABPs and their associated F-actin networks in cells, as well as the development of sophisticated in vitro reconstitutions of networks with ensembles of ABPs, have allowed the field to start uncovering the underlying principles by which cells self-organize diverse F-actin networks to execute basic cellular functions.
Collapse
Affiliation(s)
- Rachel S Kadzik
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, Illinois 60637, USA; , .,Department of Molecular BioSciences, Northwestern University, Evanston, Illinois 60208, USA;
| | - Kaitlin E Homa
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, Illinois 60637, USA; ,
| | - David R Kovar
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, Illinois 60637, USA; , .,Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, Illinois 60637, USA
| |
Collapse
|
48
|
Marchenko MA, Nefedova VV, Yampolskaya DS, Borzova VA, Kleymenov SY, Nabiev SR, Nikitina LV, Matyushenko AM, Levitsky DI. Comparative structural and functional studies of low molecular weight tropomyosin isoforms, the TPM3 gene products. Arch Biochem Biophys 2021; 710:108999. [PMID: 34339666 DOI: 10.1016/j.abb.2021.108999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/06/2021] [Accepted: 07/20/2021] [Indexed: 10/20/2022]
Abstract
Tropomyosin (Tpm) is an actin-associated protein and key regulator of actin filament structure and dynamics in muscle and non-muscle cells where it participates in many vital processes. Human non-muscle cells produce many Tpm isoforms; however, little is known yet about their structural and functional properties. In the present work, we have applied various methods to investigate the properties of five low molecular weight Tpm isoforms (Tpm3.1, Tpm3.2, Tpm3.4, Tpm3.5, and Tpm3.7), the products of TPM3 gene, which significantly differ by alternatively spliced internal exon 6 (6a or 6b) and C-terminal exon 9 (9a, 9c or 9d). Our results clearly demonstrate that the properties of these Tpm isoforms are quite different depending on sequence variations in alternatively spliced regions of their molecules. These differences can be important in further studies to explain why these Tpm isoforms play a key role in organization and dynamics of the cytoskeleton.
Collapse
Affiliation(s)
- Marina A Marchenko
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology, Russian Academy of Sciences, 119071, Moscow, Russia; Department of Biochemistry, School of Biology, Moscow State University, Moscow, 119234, Russia
| | - Victoria V Nefedova
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology, Russian Academy of Sciences, 119071, Moscow, Russia
| | - Daria S Yampolskaya
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology, Russian Academy of Sciences, 119071, Moscow, Russia
| | - Vera A Borzova
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology, Russian Academy of Sciences, 119071, Moscow, Russia
| | - Sergey Y Kleymenov
- Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, 119334, Moscow, Russia
| | - Salavat R Nabiev
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, 620049, Russia
| | - Larisa V Nikitina
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, 620049, Russia
| | - Alexander M Matyushenko
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology, Russian Academy of Sciences, 119071, Moscow, Russia
| | - Dmitrii I Levitsky
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology, Russian Academy of Sciences, 119071, Moscow, Russia.
| |
Collapse
|
49
|
Targeting the actin/tropomyosin cytoskeleton in epithelial ovarian cancer reveals multiple mechanisms of synergy with anti-microtubule agents. Br J Cancer 2021; 125:265-276. [PMID: 33981016 PMCID: PMC8292367 DOI: 10.1038/s41416-021-01420-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 04/12/2021] [Accepted: 04/22/2021] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Anti-microtubule agents are widely used to treat ovarian cancers, but the efficacy is often compromised by drug resistance. We investigated co-targeting the actin/tropomyosin cytoskeleton and microtubules to increase treatment efficacy in ovarian cancers and potentially overcome resistance. METHODS The presence of tropomyosin-3.1 (Tpm3.1) was examined in clinical specimens from ovarian cancer patients using immunohistochemistry. Combinatorial effects of an anti-Tpm3.1 compound, ATM-3507, with vinorelbine and paclitaxel were evaluated in ovarian cancer cells via MTS and apoptosis assays. The mechanisms of action were established using live- and fixed-cell imaging and protein analysis. RESULTS Tpm3.1 is overexpressed in 97% of tumour tissues (558 of 577) representing all histotypes of epithelial ovarian cancer. ATM-3507 displayed synergy with both anti-microtubule agents to reduce cell viability. Only vinorelbine synergised with ATM-3507 in causing apoptosis. ATM-3507 significantly prolonged vinorelbine-induced mitotic arrest with elevated activity of the spindle assembly checkpoint and mitotic cell death; however, ATM-3507 showed minor impact on paclitaxel-induced mitotic defects. Both combinations substantially increased post-mitotic G1 arrest with cyclin D1 and E1 downregulation and an increase of p21Cip and p27Kip. CONCLUSION Combined targeting of Tpm3.1/actin and microtubules is a promising treatment strategy for ovarian cancer that should be further tested in clinical settings.
Collapse
|
50
|
Pan X, Zhou Y, Hotulainen P, Meunier FA, Wang T. The axonal radial contractility: Structural basis underlying a new form of neural plasticity. Bioessays 2021; 43:e2100033. [PMID: 34145916 DOI: 10.1002/bies.202100033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 05/27/2021] [Accepted: 06/02/2021] [Indexed: 12/25/2022]
Abstract
Axons are the longest cellular structure reaching over a meter in the case of human motor axons. They have a relatively small diameter and contain several cytoskeletal elements that mediate both material and information exchange within neurons. Recently, a novel type of axonal plasticity, termed axonal radial contractility, has been unveiled. It is represented by dynamic and transient diameter changes of the axon shaft to accommodate the passages of large organelles. Mechanisms underpinning this plasticity are not fully understood. Here, we first summarised recent evidence of the functional relevance for axon radial contractility, then discussed the underlying structural basis, reviewing nanoscopic evidence of the subtle changes. Two models are proposed to explain how actomyosin rings are organised. Possible roles of non-muscle myosin II (NM-II) in axon degeneration are discussed. Finally, we discuss the concept of periodic functional nanodomains, which could sense extracellular cues and coordinate the axonal responses. Also see the video abstract here: https://youtu.be/ojCnrJ8RCRc.
Collapse
Affiliation(s)
- Xiaorong Pan
- Center for Brain Science, School of Life Science and Technology, Shanghaitech University, Shanghai, China
| | - Yimin Zhou
- Center for Brain Science, School of Life Science and Technology, Shanghaitech University, Shanghai, China
| | - Pirta Hotulainen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Frédéric A Meunier
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Tong Wang
- Center for Brain Science, School of Life Science and Technology, Shanghaitech University, Shanghai, China
| |
Collapse
|